WO2010108187A2 - Composés et procédés pour traiter les infections microbiennes gastro-intestinales mammaliennes - Google Patents

Composés et procédés pour traiter les infections microbiennes gastro-intestinales mammaliennes Download PDF

Info

Publication number
WO2010108187A2
WO2010108187A2 PCT/US2010/028178 US2010028178W WO2010108187A2 WO 2010108187 A2 WO2010108187 A2 WO 2010108187A2 US 2010028178 W US2010028178 W US 2010028178W WO 2010108187 A2 WO2010108187 A2 WO 2010108187A2
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
certain embodiments
aryl
heteroaryl
compounds
Prior art date
Application number
PCT/US2010/028178
Other languages
English (en)
Other versions
WO2010108187A3 (fr
Inventor
Lizbeth K. Hedstrom
Gregory D. Cuny
Deviprasad R. Gollapalli
Sivapriya Kirubakaran
Boris Striepen
Suresh Kumar Gorla
Sushi K. Maurya
Corey Robert Johnson
Mandapati Kavitha
Jihan Khan
Original Assignee
Brandeis University
University Of Georgia Research Foundation, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Brandeis University, University Of Georgia Research Foundation, Inc. filed Critical Brandeis University
Priority to US13/257,418 priority Critical patent/US8969342B2/en
Priority to EP10754239A priority patent/EP2408753A4/fr
Publication of WO2010108187A2 publication Critical patent/WO2010108187A2/fr
Publication of WO2010108187A3 publication Critical patent/WO2010108187A3/fr
Priority to US14/571,673 priority patent/US10125116B2/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C233/00Carboxylic acid amides
    • C07C233/01Carboxylic acid amides having carbon atoms of carboxamide groups bound to hydrogen atoms or to acyclic carbon atoms
    • C07C233/12Carboxylic acid amides having carbon atoms of carboxamide groups bound to hydrogen atoms or to acyclic carbon atoms having the nitrogen atom of at least one of the carboxamide groups bound to a carbon atom of a hydrocarbon radical substituted by halogen atoms or by nitro or nitroso groups
    • C07C233/15Carboxylic acid amides having carbon atoms of carboxamide groups bound to hydrogen atoms or to acyclic carbon atoms having the nitrogen atom of at least one of the carboxamide groups bound to a carbon atom of a hydrocarbon radical substituted by halogen atoms or by nitro or nitroso groups with the substituted hydrocarbon radical bound to the nitrogen atom of the carboxamide group by a carbon atom of a six-membered aromatic ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C235/00Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms
    • C07C235/02Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms having carbon atoms of carboxamide groups bound to acyclic carbon atoms and singly-bound oxygen atoms bound to the same carbon skeleton
    • C07C235/04Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms having carbon atoms of carboxamide groups bound to acyclic carbon atoms and singly-bound oxygen atoms bound to the same carbon skeleton the carbon skeleton being acyclic and saturated
    • C07C235/18Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms having carbon atoms of carboxamide groups bound to acyclic carbon atoms and singly-bound oxygen atoms bound to the same carbon skeleton the carbon skeleton being acyclic and saturated having at least one of the singly-bound oxygen atoms further bound to a carbon atom of a six-membered aromatic ring, e.g. phenoxyacetamides
    • C07C235/24Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms having carbon atoms of carboxamide groups bound to acyclic carbon atoms and singly-bound oxygen atoms bound to the same carbon skeleton the carbon skeleton being acyclic and saturated having at least one of the singly-bound oxygen atoms further bound to a carbon atom of a six-membered aromatic ring, e.g. phenoxyacetamides having the nitrogen atom of at least one of the carboxamide groups bound to a carbon atom of a six-membered aromatic ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C235/00Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms
    • C07C235/02Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms having carbon atoms of carboxamide groups bound to acyclic carbon atoms and singly-bound oxygen atoms bound to the same carbon skeleton
    • C07C235/32Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms having carbon atoms of carboxamide groups bound to acyclic carbon atoms and singly-bound oxygen atoms bound to the same carbon skeleton the carbon skeleton containing six-membered aromatic rings
    • C07C235/38Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms having carbon atoms of carboxamide groups bound to acyclic carbon atoms and singly-bound oxygen atoms bound to the same carbon skeleton the carbon skeleton containing six-membered aromatic rings having the nitrogen atom of at least one of the carboxamide groups bound to a carbon atom of a six-membered aromatic ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C237/00Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by amino groups
    • C07C237/02Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by amino groups having the carbon atoms of the carboxamide groups bound to acyclic carbon atoms of the carbon skeleton
    • C07C237/20Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by amino groups having the carbon atoms of the carboxamide groups bound to acyclic carbon atoms of the carbon skeleton the carbon skeleton containing six-membered aromatic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C255/00Carboxylic acid nitriles
    • C07C255/49Carboxylic acid nitriles having cyano groups bound to carbon atoms of six-membered aromatic rings of a carbon skeleton
    • C07C255/58Carboxylic acid nitriles having cyano groups bound to carbon atoms of six-membered aromatic rings of a carbon skeleton containing cyano groups and singly-bound nitrogen atoms, not being further bound to other hetero atoms, bound to the carbon skeleton
    • C07C255/60Carboxylic acid nitriles having cyano groups bound to carbon atoms of six-membered aromatic rings of a carbon skeleton containing cyano groups and singly-bound nitrogen atoms, not being further bound to other hetero atoms, bound to the carbon skeleton at least one of the singly-bound nitrogen atoms being acylated
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C275/00Derivatives of urea, i.e. compounds containing any of the groups, the nitrogen atoms not being part of nitro or nitroso groups
    • C07C275/28Derivatives of urea, i.e. compounds containing any of the groups, the nitrogen atoms not being part of nitro or nitroso groups having nitrogen atoms of urea groups bound to carbon atoms of six-membered aromatic rings of a carbon skeleton
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C275/00Derivatives of urea, i.e. compounds containing any of the groups, the nitrogen atoms not being part of nitro or nitroso groups
    • C07C275/28Derivatives of urea, i.e. compounds containing any of the groups, the nitrogen atoms not being part of nitro or nitroso groups having nitrogen atoms of urea groups bound to carbon atoms of six-membered aromatic rings of a carbon skeleton
    • C07C275/30Derivatives of urea, i.e. compounds containing any of the groups, the nitrogen atoms not being part of nitro or nitroso groups having nitrogen atoms of urea groups bound to carbon atoms of six-membered aromatic rings of a carbon skeleton being further substituted by halogen atoms, or by nitro or nitroso groups
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C275/00Derivatives of urea, i.e. compounds containing any of the groups, the nitrogen atoms not being part of nitro or nitroso groups
    • C07C275/28Derivatives of urea, i.e. compounds containing any of the groups, the nitrogen atoms not being part of nitro or nitroso groups having nitrogen atoms of urea groups bound to carbon atoms of six-membered aromatic rings of a carbon skeleton
    • C07C275/42Derivatives of urea, i.e. compounds containing any of the groups, the nitrogen atoms not being part of nitro or nitroso groups having nitrogen atoms of urea groups bound to carbon atoms of six-membered aromatic rings of a carbon skeleton being further substituted by carboxyl groups
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C309/00Sulfonic acids; Halides, esters, or anhydrides thereof
    • C07C309/01Sulfonic acids
    • C07C309/02Sulfonic acids having sulfo groups bound to acyclic carbon atoms
    • C07C309/03Sulfonic acids having sulfo groups bound to acyclic carbon atoms of an acyclic saturated carbon skeleton
    • C07C309/13Sulfonic acids having sulfo groups bound to acyclic carbon atoms of an acyclic saturated carbon skeleton containing nitrogen atoms, not being part of nitro or nitroso groups, bound to the carbon skeleton
    • C07C309/14Sulfonic acids having sulfo groups bound to acyclic carbon atoms of an acyclic saturated carbon skeleton containing nitrogen atoms, not being part of nitro or nitroso groups, bound to the carbon skeleton containing amino groups bound to the carbon skeleton
    • C07C309/15Sulfonic acids having sulfo groups bound to acyclic carbon atoms of an acyclic saturated carbon skeleton containing nitrogen atoms, not being part of nitro or nitroso groups, bound to the carbon skeleton containing amino groups bound to the carbon skeleton the nitrogen atom of at least one of the amino groups being part of any of the groups, X being a hetero atom, Y being any atom
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D209/00Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D209/02Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom condensed with one carbocyclic ring
    • C07D209/04Indoles; Hydrogenated indoles
    • C07D209/08Indoles; Hydrogenated indoles with only hydrogen atoms or radicals containing only hydrogen and carbon atoms, directly attached to carbon atoms of the hetero ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D209/00Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D209/02Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom condensed with one carbocyclic ring
    • C07D209/04Indoles; Hydrogenated indoles
    • C07D209/10Indoles; Hydrogenated indoles with substituted hydrocarbon radicals attached to carbon atoms of the hetero ring
    • C07D209/12Radicals substituted by oxygen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D209/00Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D209/02Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom condensed with one carbocyclic ring
    • C07D209/04Indoles; Hydrogenated indoles
    • C07D209/30Indoles; Hydrogenated indoles with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, directly attached to carbon atoms of the hetero ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D215/00Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems
    • C07D215/02Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom
    • C07D215/16Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D215/38Nitrogen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D235/00Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, condensed with other rings
    • C07D235/02Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, condensed with other rings condensed with carbocyclic rings or ring systems
    • C07D235/04Benzimidazoles; Hydrogenated benzimidazoles
    • C07D235/24Benzimidazoles; Hydrogenated benzimidazoles with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached in position 2
    • C07D235/26Oxygen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D237/00Heterocyclic compounds containing 1,2-diazine or hydrogenated 1,2-diazine rings
    • C07D237/26Heterocyclic compounds containing 1,2-diazine or hydrogenated 1,2-diazine rings condensed with carbocyclic rings or ring systems
    • C07D237/30Phthalazines
    • C07D237/32Phthalazines with oxygen atoms directly attached to carbon atoms of the nitrogen-containing ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D249/00Heterocyclic compounds containing five-membered rings having three nitrogen atoms as the only ring hetero atoms
    • C07D249/02Heterocyclic compounds containing five-membered rings having three nitrogen atoms as the only ring hetero atoms not condensed with other rings
    • C07D249/041,2,3-Triazoles; Hydrogenated 1,2,3-triazoles
    • C07D249/061,2,3-Triazoles; Hydrogenated 1,2,3-triazoles with aryl radicals directly attached to ring atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D263/00Heterocyclic compounds containing 1,3-oxazole or hydrogenated 1,3-oxazole rings
    • C07D263/52Heterocyclic compounds containing 1,3-oxazole or hydrogenated 1,3-oxazole rings condensed with carbocyclic rings or ring systems
    • C07D263/54Benzoxazoles; Hydrogenated benzoxazoles
    • C07D263/56Benzoxazoles; Hydrogenated benzoxazoles with only hydrogen atoms, hydrocarbon or substituted hydrocarbon radicals, directly attached in position 2
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D263/00Heterocyclic compounds containing 1,3-oxazole or hydrogenated 1,3-oxazole rings
    • C07D263/52Heterocyclic compounds containing 1,3-oxazole or hydrogenated 1,3-oxazole rings condensed with carbocyclic rings or ring systems
    • C07D263/54Benzoxazoles; Hydrogenated benzoxazoles
    • C07D263/56Benzoxazoles; Hydrogenated benzoxazoles with only hydrogen atoms, hydrocarbon or substituted hydrocarbon radicals, directly attached in position 2
    • C07D263/57Aryl or substituted aryl radicals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D295/00Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms
    • C07D295/16Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms acylated on ring nitrogen atoms
    • C07D295/18Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms acylated on ring nitrogen atoms by radicals derived from carboxylic acids, or sulfur or nitrogen analogues thereof
    • C07D295/182Radicals derived from carboxylic acids
    • C07D295/192Radicals derived from carboxylic acids from aromatic carboxylic acids
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D409/00Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms
    • C07D409/02Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings
    • C07D409/04Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/04Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/04Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/08Bridged systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/02Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains two hetero rings
    • C07D491/04Ortho-condensed systems
    • C07D491/044Ortho-condensed systems with only one oxygen atom as ring hetero atom in the oxygen-containing ring
    • C07D491/052Ortho-condensed systems with only one oxygen atom as ring hetero atom in the oxygen-containing ring the oxygen-containing ring being six-membered

Definitions

  • Organisms must synthesize nucleotides in order for their cells to divide and replicate. Nucleotide synthesis in mammals may be achieved through one of two pathways: the de novo synthesis pathway; or the salvage pathway. Different cell types use these pathways to differing extents.
  • Inosine-5 '-monophosphate dehydrogenase (IMPDH; EC 1.1.1.205) is an enzyme involved in the biosynthesis of guanine nucleotides.
  • IMPDH catalyzes the NAD-dependent oxidation of inosine-5 '-monophosphate (IMP) to xanthosine-5 '-monophosphate (XMP) [Jackson R. C. et. al., Nature, 256, pp. 331-333, (1975)]. Regardless of species, the reaction involves the random addition of substrates. A conserved active site Cys residue attacks the C2 position of IMP and hydride is transferred to NAD, producing NADH and the E-XMP* intermediate.
  • NADH is released and a mobile flap folds into the vacant NADH site, E-XMP* hydrolyzes and XMP is released [W. Wang and L. Hedstrom, Biochemistry 36, pp. 8479-8483 (1997); J. Digits and L. Hedstrom, Biochemistry 38, pp. 2295-2306 (1999); Gan et al, Biochemistry 42, pp 847- 863 (2003)].
  • the hydrolysis step is at least partially rate-limiting in all of the IMPDHs examined to date.
  • the enzyme is unusual in that a large conformational change occurs in the middle of a catalytic cycle.
  • IMPDH is ubiquitous in eukaryotes, bacteria and protozoa [Y. Natsumeda & S. F. Carr, Ann. N.Y. Acad., 696, pp. 88-93 (1993)].
  • the prokaryotic forms share 30-40% sequence identity with the human enzyme.
  • Each is 514 amino acids, and they share 84% sequence identity.
  • Both IMPDH type I and type II form active tetramers in solution, with subunit molecular weights of 56 kDa [Y. Yamada et. al., Biochemistry, 27, pp. 2737-27
  • IMPDH guanine nucleotides
  • B- and T-lymphocytes These cells depend on the de novo, rather than salvage pathway to generate sufficient levels of nucleotides necessary to initiate a proliferative response to mitogen or antigen [A. C. Allison et. al., Lancet II, 1179, (1975) and A. C. Allison et. al., Ciba Found. Symp., 48, 207, (1977)].
  • IMPDH is an attractive target for selectively inhibiting the immune system without also inhibiting the proliferation of other cells.
  • Immunosuppression has been achieved by inhibiting a variety of enzymes including, for example, the phosphatase calcineurin (inhibited by cyclosporin and FK-506); dihydroorotate dehydrogenase, an enzyme involved in the biosynthesis of pyrimidines (inhibited by leflunomide and brequinar); the kinase FRAP (inhibited by rapamycin); and the heat shock protein hsp70 (inhibited by deoxyspergualin).
  • the phosphatase calcineurin inhibited by cyclosporin and FK-506
  • dihydroorotate dehydrogenase an enzyme involved in the biosynthesis of pyrimidines (inhibited by leflunomide and brequinar)
  • the kinase FRAP inhibited by rapamycin
  • the heat shock protein hsp70 inhibited by deoxyspergualin
  • Inhibitors of IMPDH are also known.
  • Immunosuppressants such as MPA are useful drugs in the treatment of transplant rejection and autoimmune diseases. [R. E. Morris, Kidney Intl., 49, Suppl. 53, S-26, (1996)]. However, MPA is characterized by undesirable pharmacological properties, such as gastrointestinal toxicity and poor bioavailability. [L. M. Shaw, et. al., Therapeutic Drug Monitoring, 17, pp. 690-699, (1995)].
  • a novel noncompetitive inhibitor of !MPDH, merimepodib, bas immunosuppressive activity is orally bioavailabie, and inhibits the proliferation of primary human, mouse, rat, and dog lymphocytes at concentrations of —100 BM.
  • merimepodib is a potent, specific, and reversible (MPI)H inhibitor that selectively inhibits lymphocyte proliferation, It is currently in clinical trials to treat hepatitis C virus.
  • Nucleoside analogs such as tiazofurin, ribavirin and mizoribine also inhibit IMPDH [L. Hedstrom, et. al. Biochemistry, 29, pp. 849-854 (1990); L. Hedstrom, et al. Curr. Med. Chem. 1999, 6, 545-561]. These compounds require activation to either the adenine dinucleotide (tiazofurin) or monophosphate derivatives (ribavirin and mizoribine) that inhibit IMPDH. These activation pathways are often absent in the cell of interest. In addition, nucleoside analogs suffer from lack of selectivity and can be further metabolized to produce inhibitors of other enzymes. Therefore, nucleoside analogs are prone to toxic side effects.
  • Mycophenolate mofetil a prodrug which quickly liberates free MPA in vivo, was recently approved to prevent acute renal allograft rejection following kidney transplantation. [L. M. Shaw, et. al., Therapeutic Drug Monitoring, 17, pp. 690-699, (1995); H. W. Sollinger, Transplantation, 60, pp. 225-232 (1995)]. Several clinical observations, however, limit the therapeutic potential of this drug. [L. M. Shaw, et. al., Therapeutic Drug Monitoring, 17, pp. 690-699, (1995)]. MPA is rapidly metabolized to the inactive glucuronide in vivo. [ A. C, Allison and E. M. Eugui, Immunological Reviews, 136, pp.
  • the glucuronide then undergoes enterohepatic recycling causing accumulation of MPA in the gastrointestinal tract where it cannot exert its IMPDH inhibitory activity on the immune system. This fact effectively lowers the drug's in vivo potency, while increasing its undesirable gastrointestinal side effects.
  • IMPDH also plays a role in other physiological events. Increased IMPDH activity has been observed in rapidly proliferating human leukemic cell lines and other tumor cell lines, indicating IMPDH as a target for anti-cancer as well as immunosuppressive chemotherapy [M. Nagai et. al., Cancer Res., 51, pp. 3886-3890, (1991)]. IMPDH has also been shown to play a role in the proliferation of smooth muscle cells, indicating that inhibitors of IMPDH, such as MPA, may be useful in preventing restenosis or other hyperproliferative vascular diseases [C. R. Gregory et al., Transplantation, 59, pp.
  • Cryptosporidiosis is a severe gastrointestinal disease caused by protozoan parasites of the genus Cryptosporidium.
  • the most common causes of human disease are C. parvum and C. hominis, though disease can also result from C. felis, C. meleagridis, C. canis, and C. muris infection.
  • Small children, pregnant women, the elderly, and immuno-compromised people are at risk of severe, chronic and often fatal infection.
  • Carey, C. M., Lee, H., and Trevors, J. T., Water Res., 38, 818-62 (2004); and Fayer, R., Veterinary Parasitology, 126, 37-56 (2004) are at risk of severe, chronic and often fatal infection.
  • the Cryptosporidium parasites produce spore-like oocysts that are highly resistant to water chlorination.
  • Several large outbreaks in the U.S. have been linked to drinking and recreational water. Infection rates are extremely high, with disease manifest in 30% of exposed individuals and a 50-70% mortality rate among immuno-compromised individuals.
  • IMPDH is a key enzyme in the purine salvage pathway of C. parvum. As discussed above, IMPDH is a validated drug target in immunosuppressive, cancer and viral therapy, so the human enzymes are extremely well studied. It has recently been shown that C. parvum IMPDH has very different properties than the human enzymes and that IMPDH inhibitors block parasite proliferation in vivo [N. N. Umejiego et al, , J Biol Chem, 279 pp. 40320-40327 (2004); and B. Striepen et al, Proc Natl Acad Sci U S A, 101 pp. 3154-9 (2004)].
  • IMPDH inhibitors with improved pharmacological properties and selectivities.
  • Such inhibitors should have therapeutic potential as immunosuppressants, anti-cancer agents, anti-vascular hyperproliferative agents, antiinflammatory agents, antifungal agents, antipsoriatic and anti-viral agents.
  • selective IMPDH inhibitors that can slow or block parasite and bacterial proliferation.
  • the present invention fulfills this need and has other related advantages.
  • One aspect of the present invention relates to compounds, and pharmaceutically acceptable salts and prodrugs thereof, which are useful as inhibitors of IMPDH.
  • a compound of the invention selectively inhibits a parasitic IMPDH versus a host (e.g., mammalian) IMPDH.
  • the invention provides pharmaceutical compositions comprising one or more compounds of the invention.
  • the invention also relates to methods of treating various parasitic and bacterial infections in mammals.
  • the compounds may be used alone or in combination with other therapeutic or prophylactic agents, such as anti- virals, anti-inflammatory agents, antimicrobials and immunosuppressants.
  • Figure 1 depicts triazole compounds 1-7 and their respective IC50 values against recombinant C. parvum IMPDH.
  • Figure 2 depicts triazole compounds 8-14 and their respective IC50 values against recombinant C. parvum IMPDH.
  • Figure 3 depicts triazole compounds 15-21 and their respective IC50 values against recombinant C. parvum IMPDH.
  • Figure 4 depicts triazole compounds 22-28 and their respective IC50 values against recombinant C. parvum IMPDH.
  • Figure 5 depicts triazole compounds 29-34 and their respective IC50 values against recombinant C. parvum IMPDH.
  • Figure 6 depicts triazole compounds 35-39 and their respective IC50 values against recombinant C. parvum IMPDH.
  • Figure 7 depicts a general scheme for the preparation of various 1,2,3-triazoles.
  • Reagents and conditions: X and Y N, CH, or CCl.
  • [R 1 Me] MeCH(OH)C ⁇ CH, 0 0 C, Ph 3 P, 10 min, DEAD, rt, 12 h;
  • [R 1 H] BrCHC ⁇ CH, K 2 CO 3 , DMF, rt, 12 h;
  • Reagents and conditions (a) (i) C-PrMgBr, THF, -20 0 C, 2 h, (ii) Ph 3 P, CBr 4 , DCM, 0 0 C, 2 h; (b) 1-naphthol, K 2 CO 3 , DMF, rt, 2 h; (c) 3 M NaOH, THFiH 2 O (2:1), 80 0 C, 6 h.
  • Figure 9 depicts a representative synthetic scheme for the formation of triazoles 51.
  • Figure 11 depicts oxadiazole compound 52 and its IC50 value against recombinant C. parvum IMPDH.
  • Figure 12 depicts amide compounds 53-57 and their respective IC50 values against recombinant C. parvum IMPDH.
  • Figure 13 depicts amide compounds 58-67 and their respective IC50 values against recombinant C. parvum IMPDH.
  • Figure 14 depicts amide compounds 68-76 and their respective IC50 values against recombinant C. parvum IMPDH.
  • Figure 15 depicts amide and ester compounds 77-85 and their respective IC50 values against recombinant C. parvum IMPDH.
  • Figure 16 depicts amide compounds 86-95 and their respective IC50 values against recombinant C. parvum IMPDH.
  • Figure 17 depicts amide compounds 96-104 and their respective IC50 values against recombinant C. parvum IMPDH.
  • Figure 18 depicts amide compounds 105-113 and their respective IC50 values against recombinant C. parvum IMPDH.
  • Figure 19 depicts amide and ketone compounds 114-120 and their respective IC50 values against recombinant C. parvum IMPDH.
  • Figure 20 depicts amide compounds 121-122 and their respective IC50 values against recombinant C. parvum IMPDH.
  • Figure 21 depicts amide compounds 123-124 and their respective IC50 values against recombinant C. parvum IMPDH.
  • Figure 22 depicts amide compounds 125-129 and their respective IC50 values against recombinant C. parvum IMPDH.
  • Figure 23 depicts amide compounds 130-134 and their respective IC50 values against recombinant C. parvum IMPDH.
  • Figure 24 depicts compounds 135-140 and their respective IC 50 values against recombinant C. parvum IMPDH.
  • Figure 25 depicts amide compounds 141-145 and their respective IC50 values against recombinant C. parvum IMPDH.
  • Figure 26 depicts compounds 146-148 and their respective IC50 values against recombinant C. parvum IMPDH.
  • Figure 27 depicts amide compounds 149-152 and their respective IC50 values against recombinant C. parvum IMPDH.
  • Figure 28 depicts amide compounds 153-156 and their respective IC50 values against recombinant C. parvum IMPDH.
  • Figure 29 depicts amide compounds 157-160 and their respective IC50 values against recombinant C. parvum IMPDH.
  • Figure 30 depicts amide compounds 161-162 and their respective IC50 values against recombinant C. parvum IMPDH.
  • Figure 31 depicts amide compounds 163-167 and their respective IC50 values against recombinant C. parvum IMPDH.
  • Figure 32 depicts amide compounds 168-172 and their respective IC50 values against recombinant C. parvum IMPDH.
  • Figure 33 depicts amide compounds 173-177 and their respective IC50 values against recombinant C. parvum IMPDH.
  • Figure 34 depicts amide, ester, and ketone compounds 178-185 and their respective IC 50 values against recombinant C. parvum IMPDH.
  • Figure 35 depicts two syntheses of 188. Reagents and conditions: (a) (i) C-PrMgBr, THF, -20 0 C, 2 h, (ii) Ph 3 P, CBr 4 , DCM, 0 0 C, 2 h; (b) 1-naphthol, K 2 CO 3 , DMF, rt, 2 h; (c) 3 M NaOH, THFiH 2 O (2:1), 80 0 C, 6 h; (d) 4-chloroaniline, 0 0 C, EDCI-HCl, rt, 12 h; (e) 4- chloroaniline, cat. DMAP, DCM, rt, 2 h; (f) 4-hydroxyquinoline, K 2 CO 3 , DMF, 0 0 C, rt, 12 h.
  • Figure 37 depicts triazole compounds AlIl-Al 13 and their respective IC50 values against recombinant C. parvum IMPDH.
  • Figure 38 depicts triazole compounds Al 14-Al 19 and their respective IC 50 values against recombinant C. parvum IMPDH.
  • Figure 39 depicts amide compounds C68-C70 and their respective IC50 values against recombinant C. parvum IMPDH.
  • Figure 40 depicts amide compounds C71-C85 and their respective IC50 values against recombinant C. parvum IMPDH.
  • Figure 41 depicts amide compounds C86-C100 and their respective IC50 values against recombinant C. parvum IMPDH.
  • Figure 42 depicts phthalazinone compounds D1-D18 and their respective IC50 values against recombinant C. parvum IMPDH.
  • Figure 43 depicts phthalazinone compounds D19-D36 and their respective IC50 values against recombinant C. parvum IMPDH.
  • Figure 44 depicts phthalazinone compounds D37-D54 and their respective IC50 values against recombinant C. parvum IMPDH.
  • Figure 45 depicts phthalazinone compounds D55-D61 and their respective IC50 values against recombinant C. parvum IMPDH.
  • Figure 46 depicts pyrazole compounds N1-N18 and their respective IC50 values against recombinant C. parvum IMPDH.
  • Figure 47 depicts pyrazole compounds N19-N26 and their respective IC50 values against recombinant C. parvum IMPDH.
  • Figure 48 depicts urea compounds P1-P15 and their respective IC50 values against recombinant C. parvum IMPDH.
  • Figure 49 depicts urea compounds P16-P32 and their respective IC50 values against recombinant C. parvum IMPDH.
  • Figure 50 depicts urea compounds P33-P51 and their respective IC50 values against recombinant C. parvum IMPDH.
  • Figure 51 depicts urea compounds P52-P68 and their respective IC50 values against recombinant C. parvum IMPDH.
  • Figure 52 depicts urea compounds P69-P80 and their respective IC50 values against recombinant C. parvum IMPDH.
  • Figure 53 depicts urea compounds P81-P97 and their respective IC 50 values against recombinant C. parvum IMPDH.
  • Figure 54 depicts benzoxazole compounds Q1-Q15 and their respective IC50 values against recombinant C. parvum IMPDH.
  • Figure 55 tabulates inhibition of recombinant C. parvum IMPDH (Q7IMPDH) and human IMPDH type 2 (MMPDH2). b. ⁇ 20% inhibition at 50 ⁇ M; c. ⁇ 20% inhibition at 5 ⁇ M; d. ⁇ 10% inhibition at 5 ⁇ M; e. ⁇ 20% inhibition observed at 2 ⁇ M.
  • Figure 56 tabulates the results of metabolic and plasma stability studies on various compounds of the invention.
  • Figure 57 depicts validation of the T gondii-CpIMPDH reporter parasite. Schematics of the routes to GMP for the wild-type T gondii, T gondii-AHXGVRT , and T gondii- Q7IMPDH are shown in A, D & G respectively. Genetic studies have shown that the salvage of adenosine via adenosine kinase is the predominant route to GMP for T. gondii and IMPDH catalyzes the rate limiting step of this pathway.
  • TgHXGPRT allows for the salvage of adenosine, adenine and guanosine such that the activity of TgHXGPRT is sufficient for parasite proliferation.
  • Several transporters for the uptake of nucleobases and nucleotides have been characterized in T gondii.
  • C. parvum lacks HXGPRT and is dependent on the salvage of adenosine and thus the activity of Q?IMPDH.
  • a single adenosine transporter has been identified in the genome of C. parvum.
  • T gondii pathways shown in grey highlight the genes disrupted in the parasite clones used in this study, TgHXGRT in a previous study (HXGPRT) and TgIMPDH in this study.
  • Hyp hypoxanthine; Xan, xanthine; Gua, guanine; Guo, guanosine; Ade, adenine; Ado, adenosine; Ino, inosine; AMP, adenosine monophosphate; IMP, inosine monophosphate; XMP, xanthosine monophosphate; GMP, guanosine monophosphate; HXGPRT, hypoxanthine xanthine gunanine phosphoribosyltransferase; IMPDH, IMP dehydrogenase, 1, adenine deaminase; 2, adenosine deaminase; 3, purine nucleoside phosphorylase; 4, adenosine
  • Panels B, E & H show parasite growth in the presence of 0 ⁇ M and 7.8 ⁇ M MPA for wild-type T gondii ,T gondii-AHXGVRT , and T gondii-CpIMPDH respectively.
  • Panels C, F, and I show parasite growth curves in the presence of 0 ⁇ M and 7.8 ⁇ M MPA, with the addition of 0.33 mM xanthine to the culture media, for wild-type T. gondii, T. gondii- ⁇ HXGPRT, and T. gondii-CpIMPDH respectively. Data are representative of two independent experiments.
  • Figure 58 depicts an overview and validation of the high content imaging C. parvum growth assay.
  • A schematic representation of differential labelling of parasite and host.
  • B detail of an exemplary micrograph obtained through the screening routine. Numbers indicate object identifies after segmentation analysis.
  • Panel C shows a 2-fold titration of C. parvum oocysts where the top concentration was 1.2 xl O 6 oocysts per well.
  • panel D the ratio of the number of FITC-VVL labelled C. parvum parasites to DAPI labelled HCT-8 host cell nuclei was used to standardize each well and percent C. parvum growth (solid line) was normalized to parasites receiving DMSO alone.
  • Figure 59 depicts the identification of derivatives with high potency and selectivity in the T. gondii-CplMPDH model.
  • Panel A shows the EC 50 for a selection of compounds assayed in the T.gondii-CpIMPDH parasite model and demonstrates a range in compound selectivity and potency. Compounds were assayed in triplicate and growth inhibition was calculated on a day during the exponential phase of growth, by normalization to wells receiving DMSO alone. The EC 50 calculation was performed as described in Figure 63. Compounds A82, A89, A90, A92, A102, A103, A105, and AIlO were selected for rescreening and the mean for at least 2 replicate experiments are shown. These compounds were then tested for inhibition of C.
  • parvum panel B
  • host cell growth panel C
  • percent C. parvum growth was determined using the high-content imaging assay, with compound at 12.5 ⁇ M and 25 ⁇ M.
  • the ratio of FITC-VVL labeled C. parvum parasites to DAPI labeled HCT-8 host cell nuclei was used to standardize each well and percent growth was normalized to parasites receiving DMSO alone, the mean of triplicate wells is shown.
  • a selection of compounds were selected for re- screening and the mean over at least 2 replicate experiments is shown for compounds A90, A92, A98, A103, A105, A109 and AIlO.
  • Panel C shows percent host cell growth assayed using the pmaxGFP fluorescent HCT-8 cell line with compound at 12.5 ⁇ M and 25 ⁇ M.
  • GFP expressing HCT-8 cells were seeded at 4000 cells per well into 96-well plates and triplicate wells were spiked with test compound. Fluorescence was measured daily with a SpectraMax M22/M2e (Molecular Devices) plate reader (Ex 485, Em 530) for 7 days. Percent growth inhibition was calculated on a day during the exponential phase of growth, by normalization to wells receiving DMSO alone. A selection of compounds A89, A90 and A92 were selected for re-screening and the mean over at least two replicate experiments is shown.
  • Figure 60 depicts the correlation between Q?IMPDH enzyme inhibition and potency and selectivity in the T. gondii-CpIMPDH model.
  • p-value 0.3
  • Figure 61 depicts that compounds A103 and AIlO are potent inhibitors of C. parvum growth.
  • C. parvum growth was determined using the HCI assay. The ratio of the number of FITC-VVL labelled C. parvum parasites to DAPI labelled HCT-8 host cell nuclei was used to standardize each well and percent C. parvum growth was normalised to parasites receiving DMSO alone.
  • Panels A and B show compounds A103 and AIlO respectively (EC5o ⁇ 0.8 ⁇ M). Data shows the mean of two independent experiments with triplicate wells.
  • Figure 62 tabulates data for various compounds in enzyme assays (in the absence and presence of BSA), surrogate T. gondii model assay, host cell growth and tissue culture model of C. parvum infection.
  • N.A. not applicable; N. D., not determined, a.
  • Selectivity T. gondii- Q7IMPDH EC50 versus wild-type T. gondii EC50; b. highest concentration tested; c. Maurya et al; d. lowest concentration tested; e. Umejiego et al.; f. qPCR assay.
  • FIG 63 depicts an overview of obtaining an EC50 for T. gondii growth.
  • Fluorescent T. gondii parasites are seeded into 96-well plates and spiked with test compound. Fluorescence is measured daily with a SpectraMax M22/M2e (Molecular Devices) plate reader for 6-7 days. The fluorescence readings on a day during the exponential phase of the growth curve, for example day 4 in panel A, are used to calculated percent growth inhibition.
  • y D+(A-D)/(1 + (x/C) B ) where D is the minimum value, A is the maximum value, C is the EC50 and B is the Hill coefficient, using the SoftMax Pro v5 software, as illustrated in panel B.
  • Figure 64 shows results of various compounds in the surrogate T. gondii model.
  • Panel A shows the ECsofor a selection of compounds assayed in the T.gondii-CpIMPDH parasite model. Compounds were assayed in triplicate and growth inhibition was calculated on a day during the exponential phase of growth, by normalization to wells receiving DMSO alone. The EC 50 calculation was performed as described in figure S2. Note the highest concentration tested in panel A was for compound A30 was 20 ⁇ M.
  • Panel B shows percent host cell growth assayed using the pmaxGFP fluorescent HCT-8 cell line with compound at 25 ⁇ M and 50 ⁇ M.
  • GFP expressing HCT-8 cells were seeded at 4000 cells per well into 96-well plates and triplicate wells were spiked with test compound. Fluorescence was measured daily with a SpectraMax M22/M2e (Molecular Devices) plate reader (Ex 485, Em 530) for 7 days. Percent growth inhibition was calculated on a day during the exponential phase of growth, by normalization to wells receiving DMSO alone.
  • Figure 65 depicts the selectivity of various compounds in the surrogate T0X0/Q7IMPDH assay.
  • Figure 66 tabulates activity levels of various compounds; the surrogate Toxoplasma model is predictive for anti-Cryptosporidium activity.
  • Figure 67 tabulates the inhibition of various IMPDHs by compounds A-H. Cp, C. p ⁇ rvum; Hp, Helicobacter pylori; Bb, Borrelia burgdorferi; Sp, Streptococcus pylori; ECIMPDH S250A/L444Y, Escherichia coli IMPDH containing an alanine residue at serine240 and a leucine residue at tyrosine444. These compounds (100 ⁇ M) do not inhibit IMPDHs from E. coli, Leishmania donovanii and Tritrichomonas foetus. "Intrinsic" values (adjusted for the competition with the mobile flap) are shown in parentheses.
  • Figure 68 depicts the IMPDH reaction: a. Chemical mechanism: a conserved Cys attacks C2 of IMP and hydride is transferred to NAD + producing the covalent intermediate E- XMP*. E-XMP* is hydrolyzed with a conserved Arg residue acting as a general base to produce XMP. b. The hydride transfer reaction proceeds in an open enzyme conformation. After NADH departs, a mobile flap folds into the NAD site, carrying the catalytic Arg into the active site. Inhibitors compete with the flap, so the equilibrium between open and closed states is a determinant of inhibitor affinity . c. Phylogenetic tree of IMPDHs.
  • Figure 69 depicts that C91 inhibits H. pylori growth.
  • CFU colony forming units.
  • Filled circles DMSO alone.
  • C91 concentrations open circles, 2 ⁇ M; closed squares, 7 ⁇ M; open squares, 20 ⁇ M; closed triangles, 60 ⁇ M; open triangles, 200 ⁇ M.
  • Figure 70 depicts the x-ray crystal structure of Q?IMPDH with IMP and C64 shown from two different perspectives.
  • the electron density map prior to C64 modeling with coefficients 2Fo-Fc is contoured to l ⁇ and shown as a slate cage.
  • the electron density map prior to C64 modeling with coefficients Fo-Fc is contoured to 3 ⁇ .
  • Bromine K-edge peak anomalous dispersion map is contoured to 4 ⁇ .
  • Figure 71 depicts the C64 binding pocket of Q?IMPDH superposed with human IMPDH2. Q?IMPDH residues are labeled.
  • One aspect of the present invention relates to compounds, and pharmaceutically acceptable salts and prodrugs thereof, which are useful as inhibitors of IMPDH.
  • a compound of the invention selectively inhibits a parasitic or bacterial IMPDH versus a host (e.g., mammalian) IMPDH.
  • the present invention relates to selective inhibition of Cryptosporidium IMPDH in the presence of human inosine-5'- monophosphate-dehydrogenase (IMPDH type I and type II).
  • the invention provides pharmaceutical compositions comprising one or more compounds of the invention.
  • the invention also relates to methods of treating various parasitic and bacterial infections in mammals.
  • the compounds may be used alone or in combination with other therapeutic or prophylactic agents, such as anti-virals, anti-inflammatory agents, antimicrobials and immunosuppressants.
  • IMPDH-Mediated Diseases refers to any disease state in which the IMPDH enzyme plays a regulatory role in the metabolic pathway of that disease.
  • IMPDH-mediated disease include transplant rejection and autoimmune diseases, such as rheumatoid arthritis, multiple sclerosis, juvenile diabetes, asthma, and inflammatory bowel disease, as well as other inflammatory diseases, cancer, viral replication diseases and vascular diseases.
  • the compounds, compositions and methods of using them of the invention may be used in the treatment of transplant rejection (e.g., kidney, liver, heart, lung, pancreas (islet cells), bone marrow, cornea, small bowel and skin allografts and heart valve xenografts) and autoimmune diseases, such as rheumatoid arthritis, multiple sclerosis, juvenile diabetes, asthma, inflammatory bowel disease (Crohn's disease, ulcerative colitus), lupus, diabetes, mellitus myasthenia gravis, psoriasis, dermatitis, eczema, seborrhea, pulmonary inflammation, eye uveitis, hepatitis, Grave's disease, Hashimoto's thyroiditis, Behcet's or Sjorgen's syndrome (dry eyes/mouth), pernicious or immunohaemolytic anaemia, idiopathic adrenal insufficiency, polyglandular autoimmune syndrome, and glomerular diseases
  • IMPDH enzymes are also known to be present in bacteria, fungi, and protozoans and thus may regulate microbial growth.
  • the IMPDH-inhibitor compounds, compositions and methods described herein may be useful as antibacterials, antifungals, and/or antiprotozoans, either alone or in combination with other anti-microbial agents.
  • Microbial inhibition can be measured by various methods, including, for example, IMPDH HPLC assays (measuring enzymatic production of XMP and NADH from IMP and NAD), IMPDH spectrophotometric assays (measuring enzymatic production of NADH from NAD or XMP from IMP), IMPDH fluorometric assays (measuring enzymatic production of NADH from NAD), IMPDH radioassays (measuring enzymatic production of radiolabeled XMP from radiolabeled IMP or tritium release into water from 2- 3 H-IMP).
  • IMPDH HPLC assays measuring enzymatic production of XMP and NADH from IMP and NAD
  • IMPDH spectrophotometric assays measuring enzymatic production of NADH from NAD or XMP from IMP
  • IMPDH fluorometric assays measuring enzymatic production of NADH from NAD
  • IMPDH I and IMPDH II can be measured following an adaptation of the method described in WO 97/40028. [See, additionally, U.S. Patent Application 2004/0102497 (incorporated by reference)].
  • the inventive compounds are capable of targeting and selectively inhibiting the IMPDH enzyme in bacteria. It is known that knocking out the IMPDH gene makes some bacteria avirulent, while has no effect on others. The effectiveness probably depends on which salvage pathways are operational in a given bacteria, and the environmental niche of the infection. It has been shown that IMPDHs from H. pylori, S. pyogenes and B. burgdorferi are sensitive to the inhibitors of the invention, and that the growth of H. pylori is blocked by inhibitors of the invention.
  • Organisms belonging to these genera are responsible for illnesses such as ulcers and acid reflux (H. pylori), Lyme disease (B. burgdorferi), infection (S. pyogenes), food poisoning (C jejuni and A. butzleri), abscesses (B. capillosis), periodontitis (F. nucleatum), skin ulcers (F. nucleatum), Lemierre's syndrome (F.
  • these compounds are capable of targeting and selectively inhibiting the IMPDH enzyme in fungi, as evidenced by the mycophenolic acid sensitivity of Saccharomyces cerevisiae, Candida albicans, Cryptococcus neoformans, Aspergillus flavus and Trichophyton.
  • the inventive compounds are capable of targeting and selectively inhibiting the IMPDH enzyme in protozoans, such as Toxoplasma, Eimeria, Cryptosporidium, Plasmodium, Babesia, Theileria, Neospora, Sarcocystis, Giardia, Entamoeba, Trichomonas, Leishmania and Trypanosoma.
  • protozoans such as Toxoplasma, Eimeria, Cryptosporidium, Plasmodium, Babesia, Theileria, Neospora, Sarcocystis, Giardia, Entamoeba, Trichomonas, Leishmania and Trypanosoma.
  • these compounds are capable of targeting and selectively inhibiting the IMPDH enzyme in Cryptosporidium parvum and other Cryptosporidium species. Selected Compounds of the Invention.
  • the invention relates to a compound, or a pharmaceutically acceptable salt thereof, represented by Formula I:
  • R 1 is hydrogen, alkyl, aryl, heteroaryl, aralkyl, or heteroaralkyl
  • R 2 is hydrogen or alkyl; or R 1 and an instance of R 2 taken together with the carbon atoms to which they are attached form a 5-, 6-, or 7-membered aryl or heteroaryl ring;
  • R 3 is hydrogen or alkyl
  • Y 1 is absent, O, or NR 4 ;
  • Y 2 is absent, O, NR 4 , alkylene, -(CH 2 ) m -O-(CH 2 ) p -, -(CH 2 ) m -NR 4 -(CH 2 ) p -, -(CH 2 V
  • R 4 is hydrogen or alkyl; m is 0, 1, 2, 3, or 4; and p is 0, 1, or 2; wherein, any of the aforementioned alkyl, aryl, heteroaryl, or aralkyl may be substituted with one or more groups independently selected from the group consisting of halo, azido, alkyl, haloalkyl, aralkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, heteroaralkyl, hydroxy, alkoxy, aryloxy, heteroaryloxy, amino, nitro, sulfhydryl, imino, amido, phosphonate, phosphinate, acyl, carboxyl, alkoxycarbonyl, acyloxy, silyl, alkylthio, sulfonate, sulfonyl, sulfonamido, formyl, cyano, and isocyano.
  • the invention relates to any one of the aforementioned compounds, wherein Y 1 is O or absent.
  • the invention relates to any one of the aforementioned compounds, wherein Y 1 is O.
  • the invention relates to any one of the aforementioned compounds, wherein Y 1 is absent.
  • the invention relates to any one of the aforementioned compounds, wherein R 1 is aryl or hydrogen.
  • the invention relates to any one of the aforementioned compounds, wherein R 1 is aryl.
  • the invention relates to any one of the aforementioned compounds, wherein R 1 is phenyl. In certain embodiments, the invention relates to any one of the aforementioned compounds, wherein R 1 is hydrogen.
  • the invention relates to any one of the aforementioned compounds, wherein n is 1, 2, 3, or 4.
  • the invention relates to any one of the aforementioned compounds, wherein n is 0 or 1.
  • the invention relates to any one of the aforementioned compounds, wherein n is 1.
  • the invention relates to any one of the aforementioned compounds, wherein n is 0.
  • the invention relates to any one of the aforementioned compounds, wherein R 2 is hydrogen or alkyl.
  • the invention relates to any one of the aforementioned compounds, wherein R is hydrogen.
  • the invention relates to any one of the aforementioned compounds, wherein R is alkyl.
  • the invention relates to any one of the aforementioned compounds, wherein R 2 is methyl, ethyl, ⁇ -propyl, /-propyl, «-butyl, /-butyl, s-butyl, or t-butyl.
  • the invention relates to any one of the aforementioned compounds, wherein R 2 is methyl.
  • the invention relates to any one of the aforementioned compounds, wherein R is ethyl.
  • the invention relates to any one of the aforementioned compounds, wherein R is /-propyl.
  • the invention relates to any one of the aforementioned compounds, wherein R 1 and an instance of R 2 taken together with the carbon atoms to which they are attached form a 5- or 6-membered aryl ring.
  • the invention relates to any one of the aforementioned compounds, wherein R 1 and an instance of R 2 taken together with the carbon atoms to which they are attached form a 6-membered aryl ring. In certain embodiments, the invention relates to any one of the aforementioned compounds, wherein R 3 is hydrogen or alkyl.
  • the invention relates to any one of the aforementioned compounds, wherein R 3 is hydrogen.
  • the invention relates to any one of the aforementioned compounds, wherein Y 2 is absent.
  • the invention relates to any one of the aforementioned
  • R 5 is halo, azido, alkyl, haloalkyl, aralkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, heteroaralkyl, hydroxy, alkoxy, haloalkyloxy, aryloxy, heteroaryloxy, amino, nitro, sulfhydryl, imino, amido, phosphonate, phosphinate, acyl, carboxyl, alkoxycarbonyl, acyloxy, alkylthio, sulfonate, sulfonyl, sulfonamido, formyl, cyano, or isocyano; and q is 0 to 5 inclusive.
  • the invention relates to any one of the aforementioned
  • R 5 is selected from the group consisting of halo, alkoxy, haloalkyloxy, alkylthio, amido, and cyano.
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • R 5 is cyano
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • ⁇ - ⁇ is selected from the group consisting of hydrogen, alkyl, aryl, and heteroaryl; and R 5 is halo, azido, alkyl, haloalkyl, aralkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, heteroaralkyl, hydroxy, alkoxy, haloalkyloxy, aryloxy, heteroaryloxy, amino, nitro, sulfhydryl, imino, amido, phosphonate, phosphinate, acyl, carboxyl, alkoxycarbonyl, acyloxy, alkylthio, sulfonate, sulfonyl, sulfonamido, formyl, cyano, or isocyano; and R 6 is hydrogen or alkyl.
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to a compound, or a pharmaceutically
  • the invention relates to a compound, or a pharmaceutically
  • the invention relates to a compound, or a pharmaceutically acceptable salt thereof, represented by Formula II:
  • the invention relates to a compound, or a pharmaceutically acceptable salt thereof, represented by Formula III:
  • the invention relates to a compound, or a pharmaceutically acceptable salt thereof, represented by Formula IV:
  • the invention relates to a compound, or a pharmaceutically acceptable salt thereof, represented by Formula V:
  • the invention relates to a compound, or a pharmaceutically acceptable salt thereof, represented by Formula VI:
  • R 1 is hydrogen, alkyl, cycloalkyl, aryl, heteroaryl, aralkyl, or heteroaralkyl;
  • R 2 is hydrogen or alkyl;
  • R 3 is hydrogen or alkyl;
  • n is 0, 1, 2, 3, or 4;
  • aryl or heteroaryl is selected from the group consisting of hydrogen, alkyl, aryl, and heteroaryl; q is 0, 1, 2, 3, or 4; and
  • R 5 is halo, azido, alkyl, haloalkyl, aralkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, heteroaralkyl, hydroxy, alkoxy, haloalkyloxy, aryloxy, heteroaryloxy, amino, nitro, sulfhydryl, imino, amido, phosphonate, phosphinate, acyl, carboxyl, alkoxycarbonyl, acyloxy, alkylthio, sulfonate, sulfonyl, sulfonamido, formyl, cyano, or isocyano; wherein, any of the aforementioned alkyl, aryl, heteroaryl, or aralkyl may be substituted with one or more groups independently selected from the group consisting of halo, azido, alkyl, haloalkyl, aralkyl,
  • the invention relates to any one of the aforementioned compounds, wherein X is absent.
  • the invention relates to any one of the aforementioned compounds, wherein X is methylene.
  • the invention relates to any one of the aforementioned compounds, wherein X is -NH-.
  • the invention relates to any one of the aforementioned compounds, wherein q is 0.
  • the invention relates to any one of the aforementioned compounds, wherein q is 1.
  • the invention relates to any one of the aforementioned compounds, wherein q is 1; and R 5 is halo.
  • the invention relates to any one of the aforementioned compounds, wherein q is 1; and R 5 is bromo.
  • the invention relates to any one of the aforementioned compounds, wherein n is 1, 2, 3, or 4.
  • the invention relates to any one of the aforementioned compounds, wherein n is 1 or 2.
  • the invention relates to any one of the aforementioned compounds, wherein n is 1.
  • the invention relates to any one of the aforementioned compounds, wherein n is 2.
  • the invention relates to any one of the aforementioned compounds, wherein R 1 is hydrogen or alkyl.
  • the invention relates to any one of the aforementioned compounds, wherein R 1 is hydrogen.
  • the invention relates to any one of the aforementioned compounds, wherein R 1 is alkyl.
  • the invention relates to any one of the aforementioned compounds, wherein R 1 is methyl, ethyl, ⁇ -propyl, or /-propyl.
  • the invention relates to any one of the aforementioned compounds, wherein R is hydrogen. In certain embodiments, the invention relates to any one of the aforementioned compounds, wherein R 3 is hydrogen.
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • R 5 is amido, alkoxy, halo, haloalkyl, aryl, haloaryl, alkyl, hydroxy, alkylthio, sulfonyl, haloalkoxy, or cyano.
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • R 5 is amido, halo, or cyano.
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned and R is alky 1. In certain embodiments, the invention relates to any one of the aforementioned ; and R is methyl.
  • the invention relates to a compound, or a pharmaceutically acceptable salt thereof, selected from the group consisting of
  • the invention relates to a compound, or a pharmaceutically acceptable salt thereof, represented by Formula VII:
  • the invention relates to a compound, or a pharmaceutically acceptable salt thereof, represented by Formula VIII:
  • the invention relates to a compound, or a pharmaceutically acceptable salt thereof, represented by Formula IX:
  • R 2 is hydrogen or alkyl; m is O, 1, or 2; is aryl, heteroaryl, amino, alkyl, cycloalkyl, heterocycloalkyl, or aralkyl; wherein, any of the aforementioned alkyl, aryl, heteroaryl, or aralkyl may be substituted with one or more groups independently selected from the group consisting of halo, azido, alkyl, haloalkyl, aralkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, heteroaralkyl, hydroxy, alkoxy, aryloxy, heteroaryloxy, amino, nitro, sulfhydryl, imino, amido, phosphonate, phosphinate, acyl, carboxyl, alkoxycarbonyl, acyloxy, silyl, alkylthio, sulfon
  • the invention relates to any one of the aforementioned compounds, wherein R is hydrogen.
  • the invention relates to any one of the aforementioned compounds, wherein R 2 is alkyl.
  • the invention relates to any one of the aforementioned compounds, wherein R 2 is methyl, ethyl, n-propyl, i-propyl, n-butyl, s-butyl, i-butyl, or t-butyl. In certain embodiments, the invention relates to any one of the aforementioned compounds, wherein m is 0 or 1.
  • the invention relates to any one of the aforementioned compounds, wherein m is 0.
  • the invention relates to any one of the aforementioned compounds, wherein m is 1.
  • the invention relates to any one of the aforementioned
  • R 5 is halo, azido, alkyl, haloalkyl, aralkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, heteroaralkyl, hydroxy, alkoxy, haloalkyloxy, aryloxy, heteroaryloxy, amino, nitro, sulfhydryl, imino, amido, phosphonate, phosphinate, acyl, carboxyl, alkoxycarbonyl, acyloxy, alkylthio, sulfonate, sulfonyl, sulfonamido, formyl, cyano, or isocyano.
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • R 5 is amido, alkoxy, halo, haloalkyl, aryl, haloaryl, alkyl, hydroxy, alkylthio, sulfonyl, haloalkoxy, or cyano.
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • R 3 is amido, halo, or cyano.
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to a compound, or a pharmaceutically
  • the invention relates to a compound, or a pharmaceutically acceptable salt thereof, represented by Formula X:
  • Formula X wherein, independently for each occurrence, m is O, 1, 2, or 3; X is absent, O, S, or NH; and is aryl or heteroaryl; wherein, any of the aforementioned aryl or heteroaryl, may be substituted with one or more groups independently selected from the group consisting of halo, azido, alkyl, haloalkyl, aralkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, heteroaralkyl, hydroxy, alkoxy, aryloxy, heteroaryloxy, amino, nitro, sulfhydryl, imino, amido, phosphonate, phosphinate, acyl, carboxyl, alkoxycarbonyl, acyloxy, silyl, alkylthio, sulfonate, sulfonyl, sulfonamido, formyl, cyano, and iso
  • the invention relates to any one of the aforementioned compounds, wherein one occurrence of m is 0; and one occurrence of m is 1.
  • the invention relates to any one of the aforementioned compounds, wherein one occurrence of m is 0; and one occurrence of m is 2.
  • the invention relates to any one of the aforementioned compounds, wherein one occurrence of m is 0; and one occurrence of m is 3.
  • the invention relates to any one of the aforementioned compounds, wherein X is O.
  • the invention relates to any one of the aforementioned compounds, wherein X is S.
  • the invention relates to any one of the aforementioned compounds, wherein X is NH.
  • the invention relates to any one of the aforementioned
  • halo azido, alkyl, haloalkyl, aralkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, heteroaralkyl, hydroxy, alkoxy, haloalkyloxy, aryloxy, heteroaryloxy, amino, nitro, sulfhydryl, imino, amido, phosphonate, phosphinate, acyl, carboxyl, alkoxycarbonyl, acyloxy, alkylthio, sulfonate, sulfonyl, sulfonamido, formyl, cyano, or isocyano; and q is 0 to 5 inclusive.
  • the invention relates to any one of the aforementioned
  • R 5 is selected from the group consisting of halo, alkoxy, haloalkyloxy, alkylthio, amido, and cyano.
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • R 5 is cyano
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to a compound, or a pharmaceutically
  • the invention relates to a compound, or a pharmaceutically acceptable salt thereof, represented by Formula XI:
  • Figure XI wherein, independently for each occurrence, m is 0, 1, or 2; R 2 is hydrogen or alkyl; R 3 is hydrogen or alkyl; and is aryl or heteroaryl; wherein, any of the aforementioned alkyl, aryl, or heteroaryl may be substituted with one or more groups independently selected from the group consisting of halo, azido, alkyl, haloalkyl, aralkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, heteroaralkyl, hydroxy, alkoxy, aryloxy, heteroaryloxy, amino, nitro, sulfhydryl, imino, amido, phosphonate, phosphinate, acyl, carboxyl, alkoxycarbonyl, acyloxy, silyl, alkylthio, sulfonate, sulfonyl, sulfonamido, formyl
  • the invention relates to any one of the aforementioned compounds, wherein m is 1.
  • the invention relates to any one of the aforementioned compounds, wherein m is 1 or 2; and R 2 is hydrogen.
  • the invention relates to any one of the aforementioned compounds, wherein m is 1 or 2; and R 2 is alkyl.
  • the invention relates to any one of the aforementioned compounds, wherein m is 1 or 2; and R 2 is methyl.
  • the invention relates to any one of the aforementioned
  • R 5 is halo, azido, alkyl, haloalkyl, hydroxyalkyl, aminoalkyl, aralkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, heteroaralkyl, hydroxy, alkoxy, haloalkyloxy, aryloxy, heteroaryloxy, amino, nitro, sulfhydryl, imino, amido, phosphonate, phosphinate, acyl, carboxyl, alkoxycarbonyl, carboxylic acid, acyloxy, alkylthio, sulfonate, sulfonyl, sulfonamido, formyl, cyano, or isocyano.
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to a compound, or a pharmaceutically
  • the invention relates to a compound, or a pharmaceutically acceptable salt thereof, represented by Formula XII:
  • Formula XII wherein, independently for each occurrence, m is 0, 1, or 2; R 2 is hydrogen or alkyl; is aryl or heteroaryl; and is aryl or heteroaryl; wherein, any of the aforementioned alkyl, aryl, or heteroaryl may be substituted with one or more groups independently selected from the group consisting of halo, azido, alkyl, haloalkyl, aralkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, heteroaralkyl, hydroxy, alkoxy, aryloxy, heteroaryloxy, amino, nitro, sulfhydryl, imino, amido, phosphonate, phosphinate, acyl, carboxyl, alkoxycarbonyl, acyloxy, silyl, alkylthio, sulfonate, sulfonyl, sulfonamido, formy
  • the invention relates to any one of the aforementioned compounds, wherein m is 0.
  • the invention relates to any one of the aforementioned compounds, wherein m is 1.
  • the invention relates to any one of the aforementioned compounds, wherein m is 1 or 2; and R 2 is hydrogen.
  • the invention relates to any one of the aforementioned compounds, wherein m is 1 or 2; and R 2 is alkyl.
  • the invention relates to any one of the aforementioned
  • n p i iss n 0, 11, 27,. n orr 3 V;. n q i iss 00, 11, 27,. 3 ⁇ , n orr 44;- a anndd R R 53 is halo, azido, alkyl, haloalkyl, hydroxyalkyl, aminoalkyl, aralkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, heteroaralkyl, hydroxy, alkoxy, haloalkyloxy, aryloxy, heteroaryloxy, amino, nitro, sulfhydryl, imino, amido, phosphonate, phosphinate, acyl, carboxyl, alkoxycarbonyl, carboxylic acid, acyloxy, alkylthio, sulfonate, sulfonyl, sulfonamido, formyl, cyano, or isocyano
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • V or p is 0, 1, 2, or 3; q is 0, 1, 2, 3, or 4; and R 5 i •s halo, azido, alkyl, haloalkyl, hydroxyalkyl, aminoalkyl, heterocycloalkyl, aralkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, heteroaralkyl, hydroxy, alkoxy, haloalkyloxy, aryloxy, heteroaryloxy, amino, nitro, sulfhydryl, imino, amido, phosphonate, phosphinate, acyl, carboxyl, alkoxycarbonyl, carboxylic acid, acyloxy, alkylthio, sulfonate, sulfonyl, sulfonamido, formyl, cyano, oxime, or isocyano.
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to a compound, or a pharmaceutically
  • the invention relates to a compound, or a pharmaceutically acceptable salt thereof, represented by Formula XIII:
  • any of the aforementioned alkyl, aryl, or heteroaryl may be substituted with one or more groups independently selected from the group consisting of halo, azido, alkyl, haloalkyl, aralkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, heteroaralkyl, hydroxy, alkoxy, aryloxy, heteroaryloxy, amino, nitro, sulfhydryl, imino, amido, phosphonate, phosphinate, acyl, carboxyl, alkoxycarbonyl, acyloxy, silyl, alkylthio, sulfonate, sulfonyl, sulfonamido, formyl, cyano, and isocyano.
  • the invention relates to any one of the aforementioned compounds, wherein X is absent.
  • the invention relates to any one of the aforementioned compounds, wherein X is O.
  • the invention relates to any one of the aforementioned compounds, wherein m is 0.
  • the invention relates to any one of the aforementioned compounds, wherein m is 1.
  • the invention relates to any one of the aforementioned compounds, wherein m is l;and R 2 is hydrogen.
  • the invention relates to any one of the aforementioned compounds, wherein m is 1; and R 2 is alkyl.
  • the invention relates to any one of the aforementioned
  • R 5 is halo, azido, alkyl, haloalkyl, hydroxyalkyl, aminoalkyl, heterocycloalkyl, aralkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, heteroaralkyl, hydroxy, alkoxy, haloalkyloxy, aryloxy, heteroaryloxy, amino, nitro, sulfhydryl, imino, amido, phosphonate, phosphinate, acyl, carboxyl, alkoxycarbonyl, carboxylic acid, acyloxy, alkylthio, sulfonate, sulfonyl, sulfonamido, formyl, cyano, oxime, or isocyano.
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • R 5 is halo, azido, alkyl, haloalkyl, hydroxyalkyl, aminoalkyl, heterocycloalkyl, aralkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, heteroaralkyl, hydroxy, alkoxy, haloalkyloxy, aryloxy, heteroaryloxy, amino, nitro, sulfhydryl, imino, amido, phosphonate, phosphinate, acyl, carboxyl, alkoxycarbonyl, carboxylic acid, acyloxy, alkylthio, sulfonate, sulfonyl, sulfonamido, formyl, cyano, oxime, or isocyano.
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to any one of the aforementioned
  • the invention relates to a compound, or a pharmaceutically acceptable salt thereof, selected from the group consisting of
  • the compounds of the invention may contain one or more asymmetric carbon atoms and thus may occur as racemates and racemic mixtures, single enantiomers, diastereomeric mixtures and individual diastereomers. All such isomeric forms of these compounds are included in the present invention, unless expressly excluded.
  • Each stereogenic carbon may be of the R or S configuration.
  • the compounds of the invention described above may be modified by appending appropriate functionalities to enhance selective biological properties.
  • modifications are known in the art and include those which increase biological penetration into a given biological compartment (e.g., blood, lymphatic system, central nervous system), increase oral availability, increase solubility to allow administration by injection, alter metabolism and alter rate of excretion.
  • the invention relates to a pharmaceutical composition, comprising a pharmaceutically acceptable carrier, adjuvant, or vehicle; and any one of the aforementioned compounds.
  • the invention relates to any one of the aforementioned compositions, further comprising an antimicrobial agent. In certain embodiments, the invention relates to any one of the aforementioned compositions, further comprising an antibiotic, antifungal, or antiprotozoal agent.
  • the invention relates to any one of the aforementioned compositions, further comprising an antibiotic agent selected from the group consisting of vancomycin, metronidazole, amoxicillin, ciprofloxacin, doxycycline, gentamicin and clindamycin.
  • an antibiotic agent selected from the group consisting of vancomycin, metronidazole, amoxicillin, ciprofloxacin, doxycycline, gentamicin and clindamycin.
  • the invention relates to any one of the aforementioned compositions, further comprising an antifungal selected from the group consisting of terbinafine, flucytosine, fluconazole, itraconazole, ketoconazole, voriconazole, nikkomycin Z, caspofungin, micafungin (FK463), anidulafungin (LY303366), amphotericin B (AmB), and nystatin.
  • an antifungal selected from the group consisting of terbinafine, flucytosine, fluconazole, itraconazole, ketoconazole, voriconazole, nikkomycin Z, caspofungin, micafungin (FK463), anidulafungin (LY303366), amphotericin B (AmB), and nystatin.
  • the invention relates to any one of the aforementioned compositions, further comprising an antiprotozoal agent selected from the group consisting of eflornithine, furazolidone, melarsoprol, metronidazole, ornidazole, paromomycin sulfate, pentamidine, pyrimethamine, and tinidazole.
  • an antiprotozoal agent selected from the group consisting of eflornithine, furazolidone, melarsoprol, metronidazole, ornidazole, paromomycin sulfate, pentamidine, pyrimethamine, and tinidazole.
  • the invention relates to any one of the aforementioned compositions, further comprising an immunosuppression agent.
  • the invention relates to any one of the aforementioned compositions, further comprising an immunosuppression agent selected from the group consisting of cyclosporin A, FK506, rapamycin, lefiunomide, deoxyspergualin, prednisone, azathioprine, mycophenolate mofetil, OKT3, ATAG, interferon and mizoribine.
  • an immunosuppression agent selected from the group consisting of cyclosporin A, FK506, rapamycin, lefiunomide, deoxyspergualin, prednisone, azathioprine, mycophenolate mofetil, OKT3, ATAG, interferon and mizoribine.
  • the invention relates to any one of the aforementioned compositions, further comprising an anti-cancer agent.
  • the invention relates to any one of the aforementioned compositions, further comprising an anti-cancer agent selected from the group consisting of cis-platin, actinomycin D, doxorubicin, vincristine, vinblastine, etoposide, amsacrine, mitoxantrone, tenipaside, taxol, colchicine, cyclosporin A, phenothiazines, interferon and thioxantheres.
  • an anti-cancer agent selected from the group consisting of cis-platin, actinomycin D, doxorubicin, vincristine, vinblastine, etoposide, amsacrine, mitoxantrone, tenipaside, taxol, colchicine, cyclosporin A, phenothiazines, interferon and thioxantheres.
  • the invention relates to any one of the aforementioned compositions, further comprising an anti-viral agent. In certain embodiments, the invention relates to any one of the aforementioned compositions, further comprising an anti-viral agent selected from the group consisting of cytovene, ganciclovir, trisodium phosphonoformate, Ribavirin, d4T, ddl, AZT, and acyclovir.
  • the invention relates to any one of the aforementioned compositions, further comprising an anti-vascular hyperproliferative agent.
  • the invention relates to any one of the aforementioned compositions, further comprising an anti-vascular hyperproliferative selected from the group consisting of HMG Co-A reductase inhibitors such as lovastatin, thromboxane A2 synthetase inhibitors, eicosapentanoic acid, ciprostene, trapidil, ACE inhibitors, low molecular weight heparin, mycophenolic acid, rapamycin and 5-(3'-pyridinylmethyl)benzofuran-2-carboxylate.
  • HMG Co-A reductase inhibitors such as lovastatin, thromboxane A2 synthetase inhibitors, eicosapentanoic acid, ciprostene, trapidil, ACE inhibitors, low molecular weight heparin, mycophenolic acid, rapamycin and 5-(3'-pyridinylmethyl)benzofuran-2-carboxylate.
  • the compounds of the invention are defined to include pharmaceutically acceptable salts or prodrugs thereof.
  • a "pharmaceutically acceptable salt or prodrug” means any pharmaceutically acceptable salt, ester, salt of an ester, or other derivative of a compound of the invention which, upon administration to a recipient, is capable of providing (directly or indirectly) a compound of this invention.
  • Particularly favored prodrugs are those that increase the bioavailability of the compounds of the invention when such compounds are administered to a mammal (e.g., by allowing an orally administered compound to be more readily absorbed into the blood) or which enhance delivery of the parent compound to a biological compartment (e.g., the brain or lymphatic system) relative to the parent species.
  • Exemplary prodrugs include derivatives where a group which enhances aqueous solubility or active transport through the gut membrane is appended to the structure of the compounds of the invention.
  • Pharmaceutically acceptable salts of the compounds of the invention include those derived from pharmaceutically acceptable inorganic and organic acids and bases.
  • suitable acid salts include acetate, adipate, alginate, aspartate, benzoate, benzenesulfonate, bisulfate, butyrate, citrate, camphorate, camphorsulfonate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, formate, fumarate, glucoheptanoate, glycerophosphate, glycolate, hemisulfate, heptanoate, hexanoate, hydrochloride, hydrobromide, hydroiodide, 2-hydroxyethanesulfonate, lactate, maleate, malonate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oxalate, palmoate, pec
  • Salts derived from appropriate bases include alkali metal (e.g., sodium), alkaline earth metal (e.g., magnesium), and ammonium salts.
  • alkali metal e.g., sodium
  • alkaline earth metal e.g., magnesium
  • ammonium salts e.g., sodium
  • This invention also envisions the quaternization of any basic nitrogen-containing groups of the compounds disclosed herein. Water or oil-soluble or dispersible products may be obtained by such quaternization.
  • the invention relates to a pharmaceutical composition, wherein the pharmaceutical composition comprises any one of the aforementioned compounds or a pharmaceutically acceptable salt thereof; an additional agent selected from the group consisting of an immunosuppressant, an anti-cancer agent, an anti-viral agent, antiinflammatory agent, antifungal agent, antibiotic, and an anti-vascular hyperproliferation compound; and any pharmaceutically acceptable carrier, adjuvant or vehicle.
  • the invention relates to any one of the aforementioned pharmaceutical compositions, wherein the pharmaceutical composition comprises any one of the aforementioned compounds or a pharmaceutically acceptable salt thereof; and a pharmaceutically acceptable carrier, adjuvant or vehicle.
  • the invention relates to any one of the aforementioned pharmaceutical compositions, wherein the pharmaceutical composition optionally comprises an additional agent selected from the group consisting of an immunosuppressant, an anti-cancer agent, an anti-viral agent, antiinflammatory agent, antifungal agent, antibiotic, and an anti- vascular hyperproliferation compound.
  • an additional agent selected from the group consisting of an immunosuppressant, an anti-cancer agent, an anti-viral agent, antiinflammatory agent, antifungal agent, antibiotic, and an anti- vascular hyperproliferation compound.
  • pharmaceutically acceptable carrier or adjuvant refers to a carrier or adjuvant that may be administered to a patient, together with a compound of this invention, and which does not destroy the pharmacological activity thereof and is nontoxic when administered in doses sufficient to deliver a therapeutic amount of the compound.
  • Pharmaceutically acceptable carriers, adjuvants and vehicles that may be used in the pharmaceutical compositions of the invention include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, self-emulsifying drug delivery systems (SEDDS) such as d.alpha.
  • SEDDS self-emulsifying drug delivery systems
  • -tocopherol polyethyleneglycol 1000 succinate surfactants used in pharmaceutical dosage forms such as Tweens or other similar polymeric delivery matrices, serum proteins, such as human serum albumin, buffer substances such as phosphates, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, cellulose-based substances, polyethylene glycol, sodium carboxymethylcellulose, polyacrylates, waxes, polyethylene-polyoxypropylene-block polymers, polyethylene glycol and wool fat.
  • surfactants used in pharmaceutical dosage forms such as Tweens or other similar polymeric delivery matrices
  • serum proteins such as human serum albumin
  • buffer substances such as phosphates, glycine, sorb
  • Cyclodextrins such as ⁇ -, ⁇ -, and ⁇ -cyclodextrin, or chemically modified derivatives such as hydroxyalkylcyclodextrins, including 2- and 3-hydroxypropyl- ⁇ -cyclodextrins, or other solubilized derivatives may also be advantageously used to enhance delivery of any one of the aforementioned compounds.
  • compositions of the invention may be administered orally, parenternally, by inhalation spray, topically, rectally, nasally, buccally, vaginally or via an implanted reservoir.
  • the pharmaceutical compositions of the invention may contain any conventional non-toxic pharmaceutically-acceptable carriers, adjuvants or vehicles.
  • the pH of the formulation may be adjusted with pharmaceutically acceptable acids, bases or buffers to enhance the stability of the formulated compound or its delivery form.
  • parenternal as used herein includes subcutaneous, intracutaneous, intravenous, intramuscular, intra-articular, intraarterial, intrasynovial, intrasternal, intrathecal, intralesional and intracranial injection or infusion techniques.
  • the pharmaceutical compositions may be in the form of a sterile injectable preparation, for example, as a sterile injectable aqueous or oleaginous suspension.
  • This suspension may be formulated according to techniques known in the art using suitable dispersing or wetting agents (such as, for example, Tween 80) and suspending agents.
  • the sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally-acceptable diluent or solvent, for example, as a solution in 1,3-butanediol.
  • suitable vehicles and solvents that may be employed are mannitol, water, Ringer's solution and isotonic sodium chloride solution.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium.
  • any bland fixed oil may be employed including synthetic mono- or diglycerides.
  • Fatty acids, such as oleic acid and its glyceride derivatives are useful in the preparation of injectables, as are natural pharmaceutically-acceptable oils, such as olive oil or castor oil, especially in their polyoxyethylated versions.
  • These oil solutions or suspensions may also contain a long-chain alcohol diluent or dispersant such as those described in Pharmacopeia Helvetica, Ph.
  • HeIv. or a similar alcohol, or carboxymethyl cellulose or similar dispersing agents which are commonly used in the formulation of pharmaceutically acceptable dosage forms such as emulsions and or suspensions.
  • Other commonly used surfactants such as Tweens or Spans and/or other similar emulsifying agents or bioavailability enhancers which are commonly used in the manufacture of pharmaceutically acceptable solid, liquid, or other dosage forms may also be used for the purposes of formulation.
  • compositions of the invention may be orally administered in any orally acceptable dosage form including, but not limited to, capsules, tablets, emulsions and aqueous suspensions, dispersions and solutions.
  • carriers which are commonly used include lactose and corn starch.
  • Lubricating agents such as magnesium stearate, are also typically added.
  • useful diluents include lactose and dried corn starch.
  • compositions of the invention may also be administered in the form of suppositories for rectal administration.
  • These compositions can be prepared by mixing a compound of the invention with a suitable non-irritating excipient which is solid at room temperature but liquid at the rectal temperature and therefore will melt in the rectum to release the active components.
  • suitable non-irritating excipient include, but are not limited to, cocoa butter, beeswax and polyethylene glycols.
  • Topical administration of the pharmaceutical compositions of the invention is especially useful when the desired treatment involves areas or organs readily accessible by topical application.
  • the pharmaceutical composition should be formulated with a suitable ointment containing the active components suspended or dissolved in a carrier.
  • Carriers for topical administration of the compounds of the invention include, but are not limited to, mineral oil, liquid petroleum, white petroleum, propylene glycol, polyoxy-ethylene polyoxypropylene compound, emulsifying wax and water.
  • the pharmaceutical composition can be formulated with a suitable lotion or cream containing the active compound suspended or dissolved in a carrier with suitable emulsifying agents.
  • Suitable carriers include, but are not limited to, mineral oil, sorbitan monostearate, polysorbate 60, cetyl esters wax, cetearyl alcohol, 2-octyldodecanol, benzyl alcohol and water.
  • the pharmaceutical compositions of the invention may also be topically applied to the lower intestinal tract by rectal suppository formulation or in a suitable enema formulation. Topically-transdermal patches are also included in this invention.
  • compositions of the invention may be administered by nasal aerosol or inhalation.
  • Such compositions are prepared according to techniques well-known in the art of pharmaceutical formulation and may be prepared as solutions in saline, employing benzyl alcohol or other suitable preservatives, absorption promoters to enhance bioavailability, fiuorocarbons, and/or other solubilizing or dispersing agents known in the art.
  • Dosage levels of between about 0.01 and about 100 mg/kg body weight per day, or between about 0.5 and about 75 mg/kg body weight per day, of the IMPDH inhibitory compounds described herein are useful in a monotherapy and/or in combination therapy for the prevention and treatment of IMPDH-mediated disease or infection.
  • the pharmaceutical compositions of the invention will be administered from about 1 to about 5 times per day or alternatively, as a continuous infusion. Such administration can be used as a chronic or acute therapy.
  • the amount of active ingredient that may be combined with the carrier materials to produce a single dosage form will vary depending upon the host treated and the particular mode of administration.
  • a typical preparation will contain from about 5% to about 95% active compound (w/w). Such preparations contain from about 20% to about 80% active compound.
  • compositions of the invention comprise a combination of an IMPDH inhibitor of the invention and one or more additional therapeutic or prophylactic agents
  • both the IMPDH inhibitor and the additional agent should be present at dosage levels of between about 10 to 100%, or between about 10 to 80% of the dosage normally administered in a monotherapy regimen.
  • the additional agents may be administered separately, as part of a multiple dose regimen, from the compounds of this invention. Alternatively, those agents may be part of a single dosage form, mixed together with the compounds of the invention in a single composition.
  • a maintenance dose of a compound, composition or combination of the invention may be administered, if necessary. Subsequently, the dosage or frequency of administration, or both, may be reduced, as a function of the symptoms, to a level at which the improved condition is retained when the symptoms have been alleviated to the desired level, treatment should cease. Patients may, however, require intermittent treatment on a long-term basis upon any recurrence of disease symptoms.
  • the invention relates to a pharmaceutical composition for treatment or prevention of a protozoan infection, comprising a pharmaceutically acceptable carrier, adjuvant or vehicle and at least one of the aforementioned compounds, or a pharmaceutically acceptable salt or prodrug thereof.
  • the invention relates to any one of the aforementioned pharmaceutical compositions, wherein said protozoan infection is caused by a protozoan selected from the group consisting of the genera Toxoplasma, Eimeria, Cryptosporidium, Plasmodium, Babesia, Theileria, Neospora, Sarcocystis, Giardia, Entamoeba, Trichomonas, Tritrichomonas, Leishmania and Trypanosoma.
  • a protozoan selected from the group consisting of the genera Toxoplasma, Eimeria, Cryptosporidium, Plasmodium, Babesia, Theileria, Neospora, Sarcocystis, Giardia, Entamoeba, Trichomonas, Tritrichomonas, Leishmania and Trypanosoma.
  • the invention relates to any one of the aforementioned pharmaceutical compositions, wherein said protozoan infection is caused by a protozoan selected from the genus Cryptosporidium.
  • the invention relates to any one of the aforementioned pharmaceutical compositions, wherein said protozoan infection is caused by Cryptosporidium parvum.
  • the invention relates to any one of the aforementioned pharmaceutical compositions, wherein the pharmaceutical composition further comprises an antimicrobial agent, such as an antibiotic, antifungal, or antiprotozoal agent.
  • an antimicrobial agent such as an antibiotic, antifungal, or antiprotozoal agent.
  • antibiotic agents include, but are not limited to, vancomycin, metronidazole, amoxicillin, ciprofloxacin, doxycycline, gentamicin and clindamycin.
  • antifungal examples include, but are not limited to, terbinafine, flucytosine, fluconazole, itraconazole, ketoconazole, voriconazole, nikkomycin Z, caspofungin, micafungin (FK463), anidulafungin (LY303366), amphotericin B (AmB), and nystatin.
  • antiprotozoal agents include, but are not limited to, eflornithine, furazolidone, melarsoprol, metronidazole, ornidazole, paromomycin sulfate, pentamidine, pyrimethamine, and tinidazole.
  • the invention relates to any one of the aforementioned pharmaceutical compositions, wherein the pharmaceutical composition is used for treatment or prevention of an IMPDH-mediated disease, and comprises a pharmaceutically acceptable carrier, adjuvant or vehicle and at least one aforementioned compound.
  • the invention relates to any one of the aforementioned pharmaceutical compositions, further comprising an immunosuppression agent.
  • additional immunosuppression agents include, but are not limited to, cyclosporin A, FK506, rapamycin, leflunomide, deoxyspergualin, prednisone, azathioprine, mycophenolate mofetil, OKT3, ATAG, interferon, and mizoribine.
  • the invention relates to any one of the aforementioned pharmaceutical compositions, further comprising an anti-cancer agent.
  • anticancer agents include, but are not limited to, cis-platin, actinomycin D, doxorubicin, vincristine, vinblastine, etoposide, amsacrine, mitoxantrone, tenipaside, taxol, colchicine, cyclosporin A, phenothiazines, interferon, and thioxantheres.
  • the invention relates to any one of the aforementioned pharmaceutical compositions, further comprising an anti-viral agent.
  • anti-viral agents include, but are not limited to, cytovene, ganciclovir, trisodium phosphonoformate, Ribavirin, d4T, ddl, AZT, and acyclovir.
  • the invention relates to any one of the aforementioned pharmaceutical compositions, further comprising an anti-vascular hyperproliferative agent.
  • anti-vascular hyperproliferative agents include, but are not limited to, HMG Co-A reductase inhibitors such as lovastatin, thromboxane A2 synthetase inhibitors, eicosapentanoic acid, ciprostene, trapidil, ACE inhibitors, low molecular weight heparin, mycophenolic acid, rapamycin, and 5-(3'-pyridinylmethyl)benzofuran-2-carboxylate.
  • HMG Co-A reductase inhibitors such as lovastatin, thromboxane A2 synthetase inhibitors, eicosapentanoic acid, ciprostene, trapidil, ACE inhibitors, low molecular weight heparin, mycophenolic acid, rapamycin, and 5-(3'-pyr
  • the invention relates to any one of the aforementioned methods, wherein said microbe is a protozoon, a bacterium, or a fungus.
  • the invention relates to any one of the aforementioned methods, wherein said microbe is a protozoon or a bacterium selected from the group consisting of the genera Toxoplasma, Eimeria, Cryptosporidium, Plasmodium, Babesia, Theileria, Neospora, Sarcocystis, Giardia, Entamoeba, Trichomonas, Tritrichomonas, Leishmania, Trypanosoma, Helicobacter, Borrelia, Salmonella, Shigella, Yersinia, Streptococcus, Campylobacter, Arcobacter, Bacteroides, Fusobacterium, Burkholderia, Clostridia, Neisseria, Mycobacterium, and Acinetobacter.
  • a bacterium selected from the group consisting of the genera Toxoplasma, Eimeria, Cryptosporidium, Plasmodium, Babesia, Theileria, Neospora, Sarcocystis, Giard
  • the invention relates to any one of the aforementioned methods, wherein said microbe is a protozoon; and said protozoon is selected from the group consisting of the genera Toxoplasma, Eimeria, Cryptosporidium, Plasmodium, Babesia, Theileria, Neospora, Sarcocystis, Giardia, Entamoeba, Trichomonas, Tritrichomonas, Leishmania and Trypanosoma.
  • the invention relates to any one of the aforementioned methods, wherein said protozoon is selected from the genus Cryptosporidium.
  • the invention relates to any one of the aforementioned methods, wherein said protozoon is Cryptosporidium parvum.
  • the invention relates to any one of the aforementioned methods, wherein said microbe is a bacterium; and said bacterium is selected from the group consisting of the genera Helicobacter, Borrelia, Salmonella, Shigella, Yersinia, Streptococcus, Campylobacter, Arcobacter, Bacteroides, Fusobacterium, Burkholderia, Clostridia, Neisseria, Mycobacterium, and Acinetobacter.
  • the invention relates to a method of treating or preventing a microbial infection in a mammal, comprising the step of administering to a mammal in need thereof a therapeutically effective amount of any one of the aforementioned compounds.
  • the invention relates to a method of treating or preventing a parasitic infection in a mammal comprising the step of administering to a mammal in need thereof a therapeutically effective amount of any one of the aforementioned compounds.
  • the invention relates to any one of the aforementioned methods, wherein said microbial infection is caused by a protozoon, a bacterium, or a fungus.
  • the invention relates to any one of the aforementioned methods, wherein said microbial infection is caused by a protozoon or a bacterium selected from the group consisting of the genera Toxoplasma, Eimeria, Cryptosporidium, Plasmodium, Babesia, Theileria, Neospora, Sarcocystis, Giardia, Entamoeba, Trichomonas, Tritrichomonas, Leishmania, Trypanosoma, Helicobacter, Borrelia, Salmonella, Shigella, Yersinia, Streptococcus, Campylobacter, Arcobacter, Bacteroides, Fusobacterium, Burkholderia, Clostridia, Neisseria, Mycobacterium, and Acinetobacter.
  • a protozoon or a bacterium selected from the group consisting of the genera Toxoplasma, Eimeria, Cryptosporidium, Plasmodium, Babesia, Theileria, Neospor
  • the invention relates to any one of the aforementioned methods, wherein said microbial infection is caused by a protozoon; and said protozoon is selected from the group consisting of the genera Toxoplasma, Eimeria, Cryptosporidium, Plasmodium, Babesia, Theileria, Neospora, Sarcocystis, Giardia, Entamoeba, Trichomonas, Tritrichomonas, Leishmania and Trypanosoma.
  • the invention relates to any one of the aforementioned methods, wherein said protozoon is selected from the genus Cryptosporidium.
  • the invention relates to any one of the aforementioned methods, wherein said microbial infection is caused by Cryptosporidium parvum.
  • the invention relates to any one of the aforementioned methods, wherein said microbe is a bacterium; and said bacterium is selected from the group consisting of the genera Helicobacter, Borrelia, Salmonella, Shigella, Yersinia, Streptococcus, Campylobacter, Arcobacter, Bacteroides, Fusobacterium, Burkholderia, Clostridia, Neisseria, Mycobacterium, and Acinetobacter.
  • the invention relates to any one of the aforementioned methods, further comprising the step of co-administering to a mammal in need thereof a therapeutically effective amount of an antimicrobial agent.
  • the invention relates to any one of the aforementioned methods, wherein said antimicrobial agent is an antibiotic. In certain embodiments, the invention relates to any one of the aforementioned methods, wherein said antimicrobial agent is an antibiotic. In certain embodiments, the invention relates to any one of the aforementioned methods, wherein said antibiotic agent is selected from the group consisting of vancomycin, metronidazole, amoxicillin, ciprofloxacin, doxycycline, gentamicin, and clindamycin.
  • the invention relates to any one of the aforementioned methods, wherein said antimicrobial agent is an antifungal. In certain embodiments, the invention relates to any one of the aforementioned methods, wherein said antifungal agent is selected from the group consisting of terbinafine, flucytosine, fluconazole, itraconazole, ketoconazole, voriconazole, nikkomycin Z, caspofungin, micafungin (FK463), anidulafungin (LY303366), amphotericin B (AmB), and nystatin.
  • said antifungal agent is selected from the group consisting of terbinafine, flucytosine, fluconazole, itraconazole, ketoconazole, voriconazole, nikkomycin Z, caspofungin, micafungin (FK463), anidulafungin (LY303366), amphotericin B (AmB), and nystatin.
  • the invention relates to any one of the aforementioned methods, wherein said antimicrobial agent is an antiparasitic.
  • said antiparasitic agent is selected from the group consisting of eflornithine, furazolidone, melarsoprol, metronidazole, ornidazole, paromomycin sulfate, pentamidine, pyrimethamine, and tinidazole.
  • the invention relates to a method of treating or preventing an IMPDH-mediated disease in a mammal, comprising the step of administering to a mammal in need thereof a therapeutically effective amount of any one of the aforementioned compounds.
  • the pharmaceutical composition only comprises the IMPDH inhibitor of the invention as the active component, such methods may additionally comprise the step of administering to a mammal in need thereof a therapeutically effective amount of an agent selected from an antiinflammatory agent, immunosuppressant, an anti-cancer agent, an anti-viral agent, or an anti-vascular hyperproliferation compound.
  • additional agents may be administered to the mammal prior to, concurrently with, or following the administration of the IMPDH inhibitor composition.
  • the invention relates to any one of the aforementioned methods, wherein the IMPDH-mediated disease is transplant rejection, graft versus host disease, rheumatoid arthritis, multiple sclerosis, juvenile diabetes, asthma, inflammatory bowel disease, Crohn's disease, ulcerative colitus, lupus, diabetes, mellitus myasthenia gravis, psoriasis, dermatitis, eczema, seborrhea, pulmonary inflammation, eye uveitis, hepatitis, Grave's disease, Hashimoto's thyroiditis, Behcet's or Sjorgen's syndrome, pernicious or immunohaemolytic anaemia, idiopathic adrenal insufficiency, polyglandular autoimmune syndrome, glomerulonephritis, scleroderma, lichen planus, viteligo, autoimmune thyroiditis, alveolitis, HTLV-I, HTL V-2, HIV-I, HIV
  • the invention relates to any one of the aforementioned methods, further comprising the step of co-administering to a mammal in need thereof a therapeutically effective amount of an agent selected from the group consisting of an antiinflammatory agent, immunosuppressant, an anti-cancer agent, an anti-viral agent, and an anti-vascular hyperproliferation compound.
  • an agent selected from the group consisting of an antiinflammatory agent, immunosuppressant, an anti-cancer agent, an anti-viral agent, and an anti-vascular hyperproliferation compound.
  • the invention relates to any one of the aforementioned methods, wherein the method is useful in suppressing an immune response in a mammal.
  • diseases including, transplant rejection (e.g., kidney, liver, heart, lung, pancreas (islet cells), bone marrow, cornea, small bowel and skin allografts and heart valve xenografts), graft versus host disease, and autoimmune diseases, such as rheumatoid arthritis, multiple sclerosis, juvenile diabetes, asthma, inflammatory bowel disease (Crohn's disease, ulcerative colitus), lupus, diabetes, mellitus myasthenia gravis, psoriasis, dermatitis, eczema, seborrhea, pulmonary inflammation, eye uveitis, hepatitis, Grave's disease, Hashimoto's thyroiditis, Behcet's or Sjorgen's syndrome (dry eyes/mouth), per
  • transplant rejection e.g., kidney,
  • the invention relates to any one of the aforementioned methods, wherein the method comprises the step of administering to the mammal a composition comprising any one of the aforementioned compounds and a pharmaceutically acceptable adjuvant. In certain embodiments, the invention relates to any one of the aforementioned methods, further comprising the step of administering to a mammal in need thereof a composition comprising an additional immunosuppressant and a pharmaceutically acceptable adjuvant.
  • the invention relates to any one of the aforementioned methods, comprising the step of administering to a mammal in need thereof a composition comprising a compound of the invention; an additional immunosuppressive agent and a pharmaceutically acceptable adjuvant.
  • the invention relates to any one of the aforementioned methods, wherein the method is useful for inhibiting viral replication in a mammal.
  • Such methods are useful in treating or preventing, DNA and RNA viral diseases caused by, for example, HTLV-I and HTL V-2, HIV-I and HIV-2, nasopharyngeal carcinoma virus, HBV, HCV, HGV, yellow fever virus, dengue fever virus, Japanese encephalitis virus, human papilloma virus, rhinoviruses and Herpes viruses, such as Epstein-Barr, cytomegaloviruses and Herpes Simplex, Types 1 and 2, or Type 6. See U.S. Pat. No. 5,380,879 (incorporated by reference).
  • the invention relates to any one of the aforementioned methods, wherein the method comprises the step of administering to the mammal a composition comprising any one of the aforementioned compounds, and a pharmaceutically acceptable adjuvant. In certain embodiments, the invention relates to any one of the aforementioned methods, further comprising the step of administering to a mammal in need thereof a composition comprising an additional anti-viral agent and a pharmaceutically acceptable adjuvant.
  • the invention relates to any one of the aforementioned methods, comprising the step of administering to a mammal in need thereof a composition comprising any one of the aforementioned compounds; an additional anti-viral agent and a pharmaceutically acceptable adjuvant.
  • the invention relates to any one of the aforementioned methods, wherein the method is useful for inhibiting vascular cellular hyperproliferation in a mammal.
  • Such methods are useful in treating or preventing diseases, including, restenosis, stenosis, artherosclerosis and other hyperproliferative vascular disease.
  • the invention relates to any one of the aforementioned methods, comprising the step of administering to the mammal a composition comprising any one of the aforementioned compounds, and a pharmaceutically acceptable adjuvant. In certain embodiments, the invention relates to any one of the aforementioned methods, further comprising the step of administering to a mammal in need thereof a therapeutically effective amount of a composition comprising an additional anti-vascular hyperproliferative agent and a pharmaceutically acceptable adjuvant.
  • the invention relates to any one of the aforementioned methods, comprising the step of administering to a mammal in need thereof a therapeutically effective amount of a composition comprising any one of the aforementioned compounds; an additional anti-vascular hyperproliferative agent and a pharmaceutically acceptable adjuvant.
  • the invention relates to any one of the aforementioned methods, wherein the method is useful for inhibiting tumors and cancer in a mammal.
  • Such methods are useful in treating or preventing diseases, including, tumors and malignancies, such as lymphoma, leukemia and other forms of cancer.
  • the invention relates to any one of the aforementioned methods, comprising the step of administering to the mammal a therapeutically effective amount of a composition comprising any one of the aforementioned compounds, and a pharmaceutically acceptable adjuvant. In certain embodiments, the invention relates to any one of the aforementioned methods, further comprising the step of administering to a mammal in need thereof a therapeutically effective amount of a composition comprising an additional antitumor or anti-cancer agent and a pharmaceutically acceptable adjuvant.
  • the invention relates to any one of the aforementioned methods, comprising the step of administering to a mammal in need thereof a composition comprising any one of the aforementioned compounds; a therapeutically effective amount of an additional anti-tumor or anti-cancer agent and a pharmaceutically acceptable adjuvant.
  • the invention relates to any one of the aforementioned methods, wherein the method is useful for inhibiting inflammation and inflammatory diseases in a mammal.
  • Such methods are useful in treating or preventing diseases, including, osteoarthritis, acute pancreatitis, chronic pancreatitis, asthma and adult respiratory distress syndrome.
  • the invention relates to any one of the aforementioned methods, comprising the step of administering to the mammal a composition comprising a therapeutically effective amount of any one of the aforementioned compounds, and a pharmaceutically acceptable adjuvant. In certain embodiments, the invention relates to any one of the aforementioned methods, further comprising the step of administering to a mammal in need thereof a composition comprising a therapeutically effective amount of an antiinflammatory agent and a pharmaceutically acceptable adjuvant.
  • the invention relates to any one of the aforementioned methods, wherein the mammal is a primate, a bovine, an ovine, an equine, a porcine, a rodent, a feline, a mustelid, or a canine.
  • the invention relates to any one of the aforementioned methods, wherein the mammal is a primate.
  • the invention relates to any one of the aforementioned methods, wherein the mammal is a human. Definitions.
  • a reference to "A and/or B", when used in conjunction with open-ended language such as “comprising” can refer, in one embodiment, to A only (optionally including elements other than B); in another embodiment, to B only (optionally including elements other than A); in yet another embodiment, to both A and B (optionally including other elements); etc.
  • the phrase "at least one,” in reference to a list of one or more elements, should be understood to mean at least one element selected from any one or more of the elements in the list of elements, but not necessarily including at least one of each and every element specifically listed within the list of elements and not excluding any combinations of elements in the list of elements.
  • This definition also allows that elements may optionally be present other than the elements specifically identified within the list of elements to which the phrase "at least one" refers, whether related or unrelated to those elements specifically identified.
  • At least one of A and B can refer, in one embodiment, to at least one, optionally including more than one, A, with no B present (and optionally including elements other than B); in another embodiment, to at least one, optionally including more than one, B, with no A present (and optionally including elements other than A); in yet another embodiment, to at least one, optionally including more than one, A, and at least one, optionally including more than one, B (and optionally including other elements); etc.
  • heteroatom is art-recognized and refers to an atom of any element other than carbon or hydrogen.
  • Illustrative heteroatoms include boron, nitrogen, oxygen, phosphorus, sulfur and selenium.
  • alkyl is art-recognized, and includes saturated aliphatic groups, including straight-chain alkyl groups, branched-chain alkyl groups, cycloalkyl (alicyclic) groups, alkyl substituted cycloalkyl groups, and cycloalkyl substituted alkyl groups.
  • a straight chain or branched chain alkyl has about 80 or fewer carbon atoms in its backbone (e.g., Ci-Cgo for straight chain, C 3 -CgO for branched chain), and alternatively, about 30 or fewer.
  • cycloalkyls have from about 3 to about 10 carbon atoms in their ring structure, and alternatively about 5, 6 or 7 carbons in the ring structure.
  • fluoroalkyl denotes an alkyl where one or more hydrogens have been replaced with fluorines.
  • lower alkyl refers to an alkyl group, as defined above, but having from one to about ten carbons, alternatively from one to about six carbon atoms in its backbone structure.
  • lower alkenyl and “lower alkynyl” have similar chain lengths.
  • aralkyl is art-recognized and refers to an alkyl group substituted with an aryl group (e.g., an aromatic or heteroaromatic group).
  • alkenyl and alkynyl are art-recognized and refer to unsaturated aliphatic groups analogous in length and possible substitution to the alkyls described above, but that contain at least one double or triple bond respectively.
  • aryl is art-recognized and refers to 5-, 6- and 7-membered single-ring aromatic groups that may include from zero to four heteroatoms, for example, benzene, naphthalene, anthracene, pyrene, pyrrole, furan, thiophene, imidazole, oxazole, thiazole, triazole, pyrazole, pyridine, pyrazine, pyridazine and pyrimidine, and the like.
  • aryl groups having heteroatoms in the ring structure may also be referred to as "aryl heterocycles" or “heteroaromatics.”
  • the aromatic ring may be substituted at one or more ring positions with such substituents as described herein, for example, halogen, azide, alkyl, aralkyl, alkenyl, alkynyl, cycloalkyl, hydroxyl, alkoxyl, amino, nitro, sulfhydryl, imino, amido, phosphonate, phosphinate, carbonyl, carboxyl, silyl, alkylthio, sulfonyl, sulfonamido, ketone, aldehyde, ester, heterocyclyl, aromatic or heteroaromatic moieties, trifluoromethyl, cyano, or the like.
  • aryl also includes polycyclic ring systems having two or more cyclic rings in which two or more carbons are common to two adjoining rings (the rings are "fused rings") wherein at least one of the rings is aromatic, e.g., the other cyclic rings may be cycloalkyls, cycloalkenyls, cycloalkynyls, aryls and/or heterocyclyls.
  • ortho, meta and para are art-recognized and refer to 1,2-, 1,3- and 1,4- disubstituted benzenes, respectively.
  • the names 1 ,2-dimethylbenzene and ortho- dimethylbenzene are synonymous.
  • heterocyclyl refers to 3- to about 10-membered ring structures, alternatively 3- to about 7-membered rings, whose ring structures include one to four heteroatoms.
  • Heterocycles may also be polycycles.
  • Heterocyclyl groups include, for example, thiophene, thianthrene, furan, pyran, isobenzofuran, chromene, xanthene, phenoxanthene, pyrrole, imidazole, pyrazole, isothiazole, isoxazole, pyridine, pyrazine, pyrimidine, pyridazine, indolizine, isoindole, indole, indazole, purine, quinolizine, isoquinoline, quinoline, phthalazine, naphthyridine, quinoxaline, quinazoline, cinnoline, pteridine, carbazole, carboline, phenanthridine, acridine, pyrimidine, phenanthroline, phenazine, phenarsazine, phenothiazine, furazan, phenoxazine, pyrrolidine, o
  • the heterocyclic ring may be substituted at one or more positions with such substituents as described above, as for example, halogen, alkyl, aralkyl, alkenyl, alkynyl, cycloalkyl, hydroxyl, amino, nitro, sulfhydryl, imino, amido, phosphonate, phosphinate, carbonyl, carboxyl, silyl, alkylthio, sulfonyl, ketone, aldehyde, ester, a heterocyclyl, an aromatic or heteroaromatic moiety, trifluoromethyl, cyano, or the like.
  • substituents as described above, as for example, halogen, alkyl, aralkyl, alkenyl, alkynyl, cycloalkyl, hydroxyl, amino, nitro, sulfhydryl, imino, amido, phosphonate, phosphinate, carbonyl, carboxyl, si
  • polycyclyl or “polycyclic group” are art-recognized and refer to two or more rings (e.g., cycloalkyls, cycloalkenyls, cycloalkynyls, aryls and/or heterocyclyls) in which two or more carbons are common to two adjoining rings, e.g., the rings are "fused rings". Rings that are joined through non-adjacent atoms are termed "bridged" rings.
  • Each of the rings of the polycycle may be substituted with such substituents as described above, as for example, halogen, alkyl, aralkyl, alkenyl, alkynyl, cycloalkyl, hydroxyl, amino, nitro, sulfhydryl, imino, amido, phosphonate, phosphinate, carbonyl, carboxyl, silyl, alkylthio, sulfonyl, ketone, aldehyde, ester, a heterocyclyl, an aromatic or heteroaromatic moiety, trifluoromethyl, cyano, or the like.
  • substituents as described above, as for example, halogen, alkyl, aralkyl, alkenyl, alkynyl, cycloalkyl, hydroxyl, amino, nitro, sulfhydryl, imino, amido, phosphonate, phosphinate, carbonyl, carboxyl, silyl,
  • carrier is art-recognized and refers to an aromatic or non-aromatic ring in which each atom of the ring is carbon.
  • ring systems refers to 5 or 6 member monocyclic rings, 8, 9 and 10 membered bicyclic ring structures, and 11, 12, 13 and 14 membered tricyclic ring structures, wherein each bond in each ring may be possess any degree of saturation that is chemically feasible. When such structures contain substituents, those substituents may be at any position of the ring system, unless otherwise specified. As specified, such ring systems may optionally comprise up to 4 heteroatoms selected from N, O or S. Those heteroatoms may replace any carbon atoms in these ring systems as long as the resulting compound is chemically stable.
  • the term "monocyclic" ring system includes saturated, partially unsaturated and fully unsaturated ring structures.
  • Examples of monocyclic and bicyclic ring systems useful in the compounds of the invention include, but are not limited to, cyclopentane, cyclopentene, indane, indene, cyclohexane, cyclohexene, cyclohexadiene, benzene, tetrahydronaphthalene, decahydronaphthalene, naphthalene, pyridine, piperidine, pyridazine, pyrimidine, pyrazine, 1,2,3-triazine, 1,2,4-triazine, 1,3,5-triazine, 1,2,3,4-tetrazine, 1,2,4,5-tetrazine, 1,2,3,4- tetrahydroquinoline, quinoline, 1,2,3,4-tetrahydroisoquinoline, isoquinoline, cinnoline, phthalazine, quinazoline, quinoxaline, 1,5-naphthyridine, 1,6
  • heterocycles may be attached to the rest of the compound by any atom of the heterocycle which results in the creation of a stable structure.
  • ring atom refers to a backbone atom that makes up the ring. Such ring atoms are selected from C, N, O or S and are bound to 2 or 3 other such ring atoms (3 in the case of certain ring atoms in a bicyclic ring system).
  • ring atom does not include hydrogen.
  • nitro is art-recognized and refers to -NO 2 ;
  • halogen is art- recognized and refers to -F, -Cl, -Br or -I;
  • sulfhydryl is art-recognized and refers to -SH;
  • hydroxyl means -OH;
  • sulfonyl is art-recognized and refers to -SO 2 " .
  • Halide designates the corresponding anion of the halogens
  • pseudohalide has the definition set forth on page 560 of "Advanced Inorganic Chemistry” by Cotton and Wilkinson, that is, for example, monovalent anionic groups sufficiently electronegative to exhibit a positive Hammett sigma value at least equaling that of a halide (e.g., CN, OCN, SCN, SeCN, TeCN, N 3 , and C(CN) 3 ).
  • a halide e.g., CN, OCN, SCN, SeCN, TeCN, N 3 , and C(CN) 3 .
  • amine and “amino” are art-recognized and refer to both unsubstituted and substituted amines, e.g., a moiety that may be represented by the general formulas:
  • R51 R52 wherein R50, R51, R52 and R53 each independently represent a hydrogen, an alkyl, an alkenyl, -(CH 2 ) m -R61, or R50 and R51 or R52, taken together with the N atom to which they are attached complete a heterocycle having from 4 to 8 atoms in the ring structure;
  • R61 represents an aryl, a cycloalkyl, a cycloalkenyl, a heterocycle or a polycycle; and m is zero or an integer in the range of 1 to 8.
  • R50 and R51 (and optionally R52) each independently represent a hydrogen, an alkyl, an alkenyl, or -(CH 2 ) m -R61.
  • alkylamine includes an amine group, as defined above, having a substituted or unsubstituted alkyl attached thereto, i.e., at least one of R50 and R51 is an alkyl group.
  • acylamino is art-recognized and refers to a moiety that may be represented by the general formula:
  • R50 is as defined above, and R54 represents a hydrogen, an alkyl, an alkenyl or -(CH2)m-R61, where m and R61 are as defined above.
  • amino is art recognized as an amino-substituted carbonyl and includes a moiety that may be represented by the general formula:
  • alkylthio refers to an alkyl group, as defined above, having a sulfur radical attached thereto.
  • the "alkylthio" moiety is represented by one of -S- alkyl, -S-alkenyl, -S-alkynyl, and -S-(CH2) m -R61, wherein m and R61 are defined above.
  • Representative alkylthio groups include methylthio, ethyl thio, and the like.
  • X50 is a bond or represents an oxygen or a sulfur
  • R55 and R56 represents a hydrogen, an alkyl, an alkenyl, -(CH 2 ) m -R61or a pharmaceutically acceptable salt
  • R56 represents a hydrogen, an alkyl, an alkenyl or -(CH 2 ) m -R61, where m and R61 are defined above.
  • X50 is an oxygen and R55 or R56 is not hydrogen
  • the formula represents an "ester.”
  • X50 is an oxygen, and R55 is as defined above, the moiety is referred to herein as a carboxyl group, and particularly when R55 is a hydrogen, the formula represents a "carboxylic acid.”
  • R55 is a hydrogen
  • the formula represents a "formate.” In general, where the oxygen atom of the above formula is replaced by sulfur, the formula represents a "thiolcarbonyl" group.
  • oxime and "oxime ether” are art-recognized and refer to moieties that may be represented by the general formula:
  • R75 is hydrogen, alkyl, cycloalkyl, alkenyl, alkynyl, aryl, aralkyl, or -(CH2) m -R61.
  • the moiety is an "oxime” when R is H; and it is an "oxime ether” when R is alkyl, cycloalkyl, alkenyl, alkynyl, aryl, aralkyl, or -(CH 2 ) m -R61.
  • alkoxyl or "alkoxy” are art-recognized and refer to an alkyl group, as defined above, having an oxygen radical attached thereto.
  • Representative alkoxyl groups include methoxy, ethoxy, propyloxy, tert-butoxy and the like.
  • An "ether” is two hydrocarbons covalently linked by an oxygen. Accordingly, the substituent of an alkyl that renders that alkyl an ether is or resembles an alkoxyl, such as may be represented by one of -O-alkyl, -O-alkenyl, -O-alkynyl, -O-(CH2) m -R61, where m and R61 are described above.
  • R57 is an electron pair, hydrogen, alkyl, cycloalkyl, or aryl.
  • R50 O in which R50 and R56 are as defined above.
  • sulfamoyl is art-recognized and refers to a moiety that may be represented by the general formula:
  • sulfonyl is art-recognized and refers to a moiety that may be represented by the general formula:
  • R58 is one of the following: hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl or heteroaryl.
  • sulfoxido is art-recognized and refers to a moiety that may be represented by the general formula:
  • phosphoryl is art-recognized and may in general be represented by the formula:
  • R60 represents a lower alkyl or an aryl.
  • Analogous substitutions may be made to alkenyl and alkynyl groups to produce, for example, aminoalkenyls, aminoalkynyls, amidoalkenyls, amidoalkynyls, iminoalkenyls, iminoalkynyls, thioalkenyls, thioalkynyls, carbonyl-substituted alkenyls or alkynyls.
  • selenoalkyl is art-recognized and refers to an alkyl group having a substituted seleno group attached thereto.
  • exemplary "selenoethers" which may be substituted on the alkyl are selected from one of -Se-alkyl, -Se-alkenyl, -Se-alkynyl, and -Se-(CH 2 ) m -R61, m and R61 being defined above.
  • triflyl, tosyl, mesyl, and nonafiyl are art-recognized and refer to trifluoromethanesulfonyl, /7-toluenesulfonyl, methanesulfonyl, and nonafluorobutanesulfonyl groups, respectively.
  • trifiate, tosylate, mesylate, and nonafiate are art-recognized and refer to trifluoromethanesulfonate ester, /7-toluenesulfonate ester, methanesulfonate ester, and nonafluorobutanesulfonate ester functional groups and molecules that contain said groups, respectively.
  • each expression e.g., alkyl, m, n, and the like, when it occurs more than once in any structure, is intended to be independent of its definition elsewhere in the same structure.
  • Me, Et, Ph, Tf, Nf, Ts, and Ms represent methyl, ethyl, phenyl, trifluoromethanesulfonyl, nonafluorobutanesulfonyl, /7-toluenesulfonyl and methanesulfonyl, respectively.
  • a more comprehensive list of the abbreviations utilized by organic chemists of ordinary skill in the art appears in the first issue of each volume of the Journal of Organic Chemistry; this list is typically presented in a table entitled Standard List of Abbreviations.
  • Certain compounds contained in compositions of the present invention may exist in particular geometric or stereoisomeric forms.
  • polymers of the present invention may also be optically active.
  • the present invention contemplates all such compounds, including cis- and trans-isomers, R- and S-enantiomers, diastereomers, (D)-isomers, (L)- isomers, the racemic mixtures thereof, and other mixtures thereof, as falling within the scope of the invention. Additional asymmetric carbon atoms may be present in a substituent such as an alkyl group. All such isomers, as well as mixtures thereof, are intended to be included in this invention.
  • a particular enantiomer of compound of the present invention may be prepared by asymmetric synthesis, or by derivation with a chiral auxiliary, where the resulting diastereomeric mixture is separated and the auxiliary group cleaved to provide the pure desired enantiomers.
  • the molecule contains a basic functional group, such as amino, or an acidic functional group, such as carboxyl, diastereomeric salts are formed with an appropriate optically-active acid or base, followed by resolution of the diastereomers thus formed by fractional crystallization or chromatographic means well known in the art, and subsequent recovery of the pure enantiomers.
  • substitution or “substituted with” includes the implicit proviso that such substitution is in accordance with permitted valence of the substituted atom and the substituent, and that the substitution results in a stable compound, e.g., which does not spontaneously undergo transformation such as by rearrangement, cyclization, elimination, or other reaction.
  • the term "substituted" is also contemplated to include all permissible substituents of organic compounds.
  • the permissible substituents include acyclic and cyclic, branched and unbranched, carbocyclic and heterocyclic, aromatic and nonaromatic substituents of organic compounds.
  • Illustrative substituents include, for example, those described herein above.
  • the permissible substituents may be one or more and the same or different for appropriate organic compounds.
  • the heteroatoms such as nitrogen may have hydrogen substituents and/or any permissible substituents of organic compounds described herein which satisfy the valences of the heteroatoms. This invention is not intended to be limited in any manner by the permissible substituents of organic compounds.
  • the chemical elements are identified in accordance with the Periodic Table of the Elements, CAS version, "Handbook of Chemistry and Physics", 67th Ed., 1986-87, inside cover.
  • treating refers to the alleviation of symptoms of a particular disorder in a patient or the improvement of an ascertainable measurement associated with a particular disorder.
  • patient refers to a mammal, including a human.
  • the resulting material (360 mg, 1.58 mmol) was dissolved in anhydrous THF and cooled to -78 0 C. Next, n-BuLi (121 mg, 1.90 mmol) was gradually added and the resulting solution stirred for 2 h at -78 0 C. The mixture was quenched with water (50 mL), allowed to stir at room temperature for 30 min, and then extracted with ethyl acetate (3 x 100 mL).
  • reaction mixture was allowed to stir at room temperature for 10 min and then finely powdered CuI (194.2 mg, 1.02 mmol) was added portion wise. After 30 min of stirring at room temperature, the reaction mixture was quenched with saturated aqueous NH 4 Cl, diluted with water (50 mL) and extracted with chloroform (3 x 50 mL). The combined organic extracts were washed with brine, dried over anhydrous MgSO 4 , filtered, concentrated in vacuo and purified by flash column chromatography using ethyl acetate/r ⁇ -hexane (a gradient of 10-20%) to furnish 3 (167 mg, 87 %) as a gelatinous solid.
  • N-(4-chlorophenyl)-2-cyclopropyl-2-(l- naphthalenyloxy)acetamide (119): To a solution of 2-cyclopropyl-2-(l- naphthalenyloxy)acetic acid (120 mg, 0.49 mmol) and 4-chloroaniline (44.0 ⁇ L, 0.49 mmol) in anhydrous DCM (10 mL) under N 2 cooled at 0 0 C was added EDCI-HCl (187.9 mg, 0.98 mmol) portion wise. The resulting solution was stirred at room temperature for 12 h.
  • reaction mixture was diluted with water (50 mL) and extracted with ethyl acetate (3 x 100 mL). The organic extracts were combined, washed with brine, dried over anhydrous MgSO 4 , filtered, and concentrated in vacuo.
  • Example 35 Heliobacter pylori, Borrelia burgdorferi, and Streptococcus pyogenes IMPDH screen
  • IC50 values of various compounds of the invention were determined for recombinant IMPDHs from H. pylori, B. burgdorferi, and S. pyogenes.
  • Example 36 Structural Basis of Cryptosporidium-Specific IMP Dehydrogenase Inhibitor Selectivity
  • Cryptosporidium parvum is a potential bio-warfare agent, an important AIDS pathogen and a major cause of diarrhea and malnutrition.
  • This parasite relies on inosine 5 '-monophosphate dehydrogenase (IMPDH) to obtain guanine nucleotides and inhibition of this enzyme blocks parasite proliferation.
  • IMPDH inosine 5 '-monophosphate dehydrogenase
  • Cryptosporidium spp. are a major cause of the "vicious cycle" of diarrhea and malnutrition in the developing world and a potential bioterrorism agent. This disease is prolonged and life-threatening in immuno-compromised patients.
  • IMPDH inosine 5 '-monophosphate dehydrogenase
  • the gene encoding Q?IMPDH appears to have been obtained from a bacteria via lateral gene transfer; we have exploited this unexpected divergence of parasite and host enzymes to identify Q?IMPDH-specific inhibitors in a high throughput screen.
  • IMPDH inosine 5 '-monophosphate dehydrogenase
  • Recombinant Q?IMPDH was purified as described previously and crystallized using the hanging drop vapor diffusion method.
  • Protein solution (4 mg/mL IMPDH, 50 mM Tris-HCl, pH 7.5, 150 mM KCl, 5% glycerol and 2 mM DTT) was mixed with well solution (34% PEG 4000, 25 mM sodium acetate and 100 mM Tris-HCl, pH 8.5) in a 1:1 ratio.
  • Data were collected from a single crystal at IOOK at beamline 8-BM at Advanced Photon Source (Argonne National Laboratory, Argonne, IL).
  • the crystals had the symmetry of space group P2i2i2.
  • the asymmetric unit contains one tetramer, which is the active form of IMPDH.
  • C64 binds in an unprecedented fashion.
  • Inhibitors of human IMPDH2 such as mycophenolic acid and merimepodib bind in the nicotinamide subsite, stacking against the purine ring of IMP in a parallel fashion, and extend either into the NAD site or into a pocket adjacent the active site but within the same monomer.
  • the thiazole ring of C64 stacks against the purine ring of IMP perpendicularly, and the remainder of C64 extends across the subunit interface into a pocket in the adjacent monomer, where the bromoaniline moiety interacts with Tyr358' (where ' denotes a residue from the adjacent subunit; Figure 71).
  • This residue forms a hydrogen bonding network involving Glu329, Ser354, Thr221 and possibly the amide nitrogen of C64 ( Figure 71).
  • Ser22', Pro26', Alal65, Gly357' form the remainder of the inhibitor binding pocket. With the exception of Thr221, all of these residues are different in human IMPDHs ( Figure 71). Thus these interactions account for the selectivity of C64 for CjpIMPDH over human IMPDHs.
  • the Q?IMPDH inhibitory activity of the compounds was assessed by monitoring the production of NADH by fluorescence (Figure 55).
  • Replacing the p-MeO of the parent compound C with Cl or Br increased potency by 10-fold (ClO, aka 126) and 20-fold (C14, aka 130), respectively, as has been similarly observed with another inhibitor series.
  • the /? ⁇ r ⁇ -substituted aniline group was replaced with 3,4-dichloroaniline (C86) or 2-naphthylamine (C90); the addition of a second Cl improved potency by a factor of 2, while fusing an additional aromatic ring increased potency by a factor of 8.
  • This molecule has demonstrated uncompetitive inhibition with respect to IMP and noncompetitive (mixed) inhibition with respect to NAD + . It was also shown to bind the nicotinamide subsite and to directly or indirectly impose on the ADP site.
  • SAR structure-activity relationship
  • Benzimidazole analogs were synthesized following the procedure outlined in Figure 55.
  • Various acetylamide derivatives ( Figure 55, 3) were prepared by treating substituted anilines (Figure 55, 1) with bromo acetylchloride (Figure 55, 2) in dichloromethane (DCM) and in the presence of catalytic amounts of N, N-dimethylaminopyridine (DMAP).
  • Various 2- substituted benzimidazoles Figure 55, 6) were prepared by condensing O-phenylene diamine ( Figure 55, 4) with aromatic aldehydes followed by oxidation in the presence of sodium metabisulfite.
  • 2-substituted benzimidazoles were coupled with the acetylamides ( Figure 55, 3) in the presence of potassium carbonate to yield derivatives of C.
  • Phenyl (C17, compound 133), pyridyl (C16, compound 132) rings are also tolerated in the 2-position.
  • Substituted phenyls (C31 (compound 147), C59 (compound 169)) are not as active as the non-substituted.
  • the potent inhibitor C64 (compound 174) has been co-crystallized with Cp- IMPDH.
  • the co-crystallized structure of Q7-IMPDH with C64 is solved and the SAR matches perfectly well with the structure.
  • 2-aromatic substitution in the benzimidazole portion is important since it interacts with the IMP in the active site according to the crystal structure.
  • the amide bond is also very important for the activity since it could potentially form hydrogen bonding with the active site residues.
  • T gondii reporter parasite suitable for the screening of Q)IMPDH inhibitors.
  • T gondii reporter parasite that mirrors the Cryptosporidium purine metabolism.
  • Figure 57 summarizes the main differences between the two parasites in this pathway.
  • T. gondii knockout mutant that, like C. parvum, lacks the ability to salvage xanthine and guanine via HXGPRT (T gor ⁇ ft ⁇ - ⁇ HXGPRT(lO)) and introduced the CpIMPDH gene under the control of a T gondii promoter.
  • T gondii IMPDH was disrupted by replacing the entire coding sequence with a chloramphenicol acetyl transferase cassette using a new cosmid-based gene targeting approach. Successful disruption of the gene was confirmed by PCR and Southern blotting; note that numerous attempts by independent laboratories failed to target this locus using smaller plasmid-based constructs. This manipulation created strain T go «Jzz-Q?IMPDH- ⁇ HXGPRT- ⁇ rgIMPDH. Lastly, we introduced a fluorescent protein cassette and isolated stable transgenic parasites by cell sorting. The resulting strain is referred to as T. gondii-CpIMPDH.
  • gondii- ⁇ XG ⁇ RT are sensitive to MPA ( Figure 57), but T. gondii-CplMPOH is resistant ( Figure 57).
  • Supplementation of the media with xanthine (0.33 mM) essentially renders wild-type T. gondii MPA resistant (EC50 ⁇ 78 ⁇ M), but has no effect on T. gondii-AHXGPRT ( Figure 57).
  • the MPA EC 50 >65 ⁇ M for T. gondii- Q7IMPDH is significantly higher, as expected given the resistance of the prokaryotic Q7IMPDH, and is independent of xanthine supplementation ( Figure 57).
  • T. gondii model system provides a powerful tool for the evaluation of in vivo efficacy, selectivity, and specificity of Q?IMPDH inhibitors.
  • Compounds that selectively inhibit Q?IMPDH will block the proliferation of T. gondii- Q7IMPDH but not the wild-type and T. gondii-AHXGPRT strains that depend on an enzyme much like the human host.
  • non-specific compounds that have off-target activities in the parasite or the host cell will inhibit the growth of all three strains.
  • a general non-selective inhibitor of both prokaryotic and eukaryotic IMPDH inhibitors will block the proliferation of both T. gondii-CplMPOH and T.
  • gondii- ⁇ HXGPRT but will have no effect on the wild-type strain; note that such compounds should be detected in our enzyme assays and eliminated before they reach this screen.
  • compounds showing poor efficacy against the T. gondii-CplMPDH parasite may signal problems pertaining to compound uptake, stability or metabolism. Examples of these varied outcomes are discussed below.
  • Figure 58 shows an overview of the methodology. Plates are fixed, permeabilized and stained with FITC- WL and DAPI to numerate parasites and host cells, respectively. Using a spinning disc microscope, we imaged a X mm area of each well, providing a robust sample typically consisting of -6000 host cells and -2000 parasite stages. The instrument is programmed to automatically move from well to well, focus and acquire 20 ⁇ M deep image stacks for the entire plate.
  • a series of automated image compression, manipulation, and object- finding algorithms was optimized for the recognition of host cells and parasites using the DAPI and FITC channels.
  • control wells are included for background subtraction.
  • the massive data output is stored, managed and accessed through an Accelrys pipeline database that performs further statistical analyses and transforms raw counts into percentage growth relative to a "no drug" control.
  • Figure 58 shows a 2-fold titration of oocysts where the highest inoculum as 1.2xlO 6 oocysts per well, for C. parvum growth assays 5xlO 5 oocysts were added per well.
  • the T. gondii model and predicting off-target host cell effects were also assayed to assess the contribution of host cell effects to antiparasitic activity ( Figures 59, 62, and 64).
  • strong host cell effects are observed in compounds that display little selectivity in the T. gondii model ( Figures 59, 62, and 64).
  • compounds that inhibited the proliferation of wild-type T. gondii with EC50 ⁇ 10 ⁇ M also inhibited the proliferation of host cells.
  • Three compounds (A82, A90, and A105) display little selectivity in the T.
  • gondii model and do not inhibit host cell growth, suggesting that the antiparasitic activities of A82, A90, and A105 do not result from the inhibition of Q7IMPDH or TgIMPDH. Instead, A82, A90, and A105 may act on other T. gondii targets not present in the host cell.
  • AlOO, A102, and A103 have EC50 >20 ⁇ M against wild- type T. gondii yet inhibit HCT-8 cell growth significantly at 12.5 ⁇ M and 25 ⁇ M ( Figure 62).
  • the average area of the host cell nucleus was also recorded as a potential indicator of host cell cytotoxicity and likewise no significant change in host cell nuclei size was detected (data not shown). Encouragingly there was a negative trend between anticryptosporidial activity and host cell growth inhibition (data not shown), indicating that improvements in anticryptosporidial activity are not coincident with secondary effects on the host cell.
  • T. gondii-CpIMPDH While not every compound that showed activity against the T. gondii-CpIMPDH parasite had strong anticryptosporidial activity, none of the compounds showing poor activity in the T. gondii-CpIMPDH model display significant anticryptosporidial activity.
  • the T. gondii assay also immediately flagged compounds with poor bioavailability and those that showed parasite killing due to off-target effects.
  • We conclude that the T. gondii-CplMPOH model provides valuable information regarding compound specificity and is a fast and highly informative filter for compound progression through medicinal chemistry optimization.
  • T. gondii model parasite that mirrors Cryptosporidium purine nucleotide pathways and depends on Q?IMPDH.
  • the T. gondii model reliably eliminates compounds from further consideration and provides a useful filter to identify off-target activities.
  • efficacy in the T. gondii model does not always guarantee anti-cryptosporidial activity.
  • T. gondii and C. parvum infect different tissues, and occupy different intracellular compartments.
  • the paras itophorous membrane of T. gondii is in direct contact with the host cell cytoplasm.
  • C. parvum remains beneath the apical membrane of the host cell and is considered 'extracytoplasmic' due to the presence of a parasite induced host cell actin patch along with other peculiar and still largely uncharacterized structures including a dense band visible in electron micrographs. This band separates the parasite's paras itophorous vacuole from the host cell cytoplasm and has been hypothesized to be involved in drug and nutrient uptake.
  • the two parasites, and their respective host cells have different repertoires of drug efflux transporters, which can also account for the differences in inhibitor sensitivity.
  • T. gondii assay does not fully negate the necessity of testing in Cryptosporidium directly, it has proven indispensible to winnow candidate compounds to a manageable number amenable to this more challenging model.
  • Al 03 and AIlO Two promising candidates for anticryptosporidial chemotherapy: Al 03 and AIlO. These compounds are >100x more potent than paromomycin, the current standard for anticryptosporidial activity.
  • HCT-8 human ileocecal adenocarcinoma epithelial cell line
  • HCT-8 cells were maintained in RPMI- 1640 (Hyclone) supplemented with 10% FBS, 1 mM sodium pyruvate, 50 U/m penicillin, 50 ⁇ g/mL streptomycin, and amphotericin B.
  • Cryptosporidium parvum oocysts were a kind gift from either Dr. Mead (Emory University) of Dr Kissinger (University of Georgia). Purified oocysts were received in 2% potassium dichromate and stored at 4 0 C for up to 4 months.
  • This oocyst suspension was diluted directly with DMEM (Hyclone) supplemented with 2% FBS, 50 U/m penicillin, 50 ⁇ g/mL streptomycin, amphotericin B and 0.2 mM L-glutamine (infection medium) to inoculate host cell monolayers at 5 x 10 5 oocysts per well.
  • Oocysts were cultured on host cell monolayers for 3 hours at 37 0 C.
  • Unexcysted oocysts and oocyst walls were then removed by aspiration and each well washed with 0.2 mL PBS (pH7.2). Infection medium was then added to the monolayers and infection was allowed to progress for 48 hours.
  • Vicia villosa lectin (WL) immunofluorescence assay and High Content Imaging Vicia villosa lectin (WL) immunofluorescence assay and High Content Imaging.
  • the WL IFA was performed in a 96-well format as follows. Following 48 hours of culture C. parvum infected HCT-8 monolayers were washed with 0.2 mL/well PBS and the monolayer was fixed with 0.2 mL/well of 3% paraformaldehyde/PBS, permeabilized with 0.25% Triton- X-100/PBS and blocked with 4% BS A/PBS. When necessary plates were stored at 4 0 C for up to 2 weeks.0.1 mL of fluorescein (FITC)-conjugated WL (Vector Labs) at 0.5 ⁇ g/mL in 1% BSA/PBS was applied to wells and incubated for 45 minutes.
  • FITC fluorescein
  • the plates were washed twice with 200 ⁇ L/well of PBS, in the first wash DAPI at 0.1 ⁇ g/mL was included. Finally 200 ⁇ L/well of PBS was added to the plates prior to storage at 4 0 C protected from light.
  • test compounds were stored as 0.1 M stocks in DMSO at -20 0 C and further diluted in DMSO to a 200X working stocks for each dilution, such that the final concentration of DMSO in the infection medium was 0.5%.
  • DMSO no drug control
  • DMSO alone was added to triplicate wells.
  • a high paromomycin concentration (0.8 mg/mL) was included on each plate in triplicate wells. Plates where this paromomycin control did not inhibit 70-80% of parasite growth were manually inspected to confirm appropriate imaging and analysis. Plates were omitted from final analysis where it was apparent that a lack of inhibition was due to poor parasite growth.
  • HCT-8 host cell growth assay HCT-8 cells were transfected with the pmaxGFP plasmid (Amaxa) using Lipofectamine (Invitrogen) following the manufactures instruction.Fluorescent lines were then selected and cloned using FACS. Confluent monolayers of pmaxGFP expressing cells were harvested from T75 flasks and passed through a 40 ⁇ m cell strainer. Cells were then seeded at 4000 cells per well in a volume of 200 ⁇ L into black, optical quality, thin bottom, 96-well plates (DB Falcon). All test compounds were diluted in DMSO to prepare a 200X working stock for each dilution.
  • the protozoan parasite Cryptosporidium parvum is a major cause of gastrointestinal disease; no effective drug treatment exists to treat this infection.
  • Q?IMPDH is most closely related to prokaryotic IMPDHs, suggesting that the parasite obtained its IMPDH gene via horizontal transfer.
  • inhibitors of Q?IMPDH that do not inhibit human IMPDHs.
  • these compounds also inhibit IMPDHs from Helicobacter pylori, Borrelia burgdorferi, and Streptococcus pyogenes, but not IMPDHs from Escherichia coli, Tritrichomonas foetus and Leishmania donovani.
  • a second generation inhibitor blocks H pylori growth.
  • IMPDH-targeted inhibitors represent a new class of antibiotics for treatment of a wide variety of pathogenic bacteria, including extensively drug resistant strains.
  • CpIMPDH is most closely related to HpIMPDH ( Figure 68), but also has -50% sequence identity to EcIMPDH, BbIMPT)H and SpIMPDH. Sequence identity drops to 32% for JjIMPDH, which is comparable to that of the eukaryotic enzymes.
  • EcIMPDH, BbIMPOH, JjIMPDH, SpIMPDH and ZdIMPDH have been characterized previously.
  • the kinetic parameters of HpIMPDH are very similar to those of CpIMPDH, and are generally characteristic of bacterial IMPDHs. Importantly, structures are available for J)IMPDH, SpIMPDH and BbIMPOH as well as for QrtMPDH and the human enzymes.
  • the compounds also inhibit BbIMPOH with similar potency to CpIMPDH and HpIMPDH ( Figure 67).
  • G and H are submicromolar inhibitors of SpIMPOH
  • A-F are markedly less effective against this enzyme, with IC50 values ranging from 13 to 90 ⁇ M.
  • No inhibition of EcIMPDH and J)IMPDH is observed at 100 ⁇ M, indicating that the values of IC 50 for A-H must be >1000 ⁇ M. This result is especially surprising for EcIMPDH because this enzyme has the same overall similarity to Q?IMPDH as the sensitive enzymes.
  • A-H do not inhibit LdIMPDH.
  • H. pylori Inhibition of H. pylori growth.
  • H. pylori is cultured in a nutrient rich medium ⁇ Brucella broth), which provides a stringent test for the antibiotic potential of IMPDH-targeted inhibitors.
  • Figure 69 shows that 20 ⁇ M C91 is sufficient to block the proliferation of a H. pylori culture exiting stationary phase. Higher concentrations of C91 display bacteriocidal effects, with only 23% of the colony forming units remaining after 24 hr treatment with 200 ⁇ M. Exponentially growing H. pylori cells are also sensitive to C91; a concentration of 60 ⁇ M is sufficient to block growth while higher concentrations are bacteriocidal.
  • Tiazofurin inhibition illustrates the magnitude of the conformational contribution to inhibitor selectivity.
  • the tiazofurin binding site is conserved among prokaryotic IMPDHs, which predicts that QrtMPDH, HpIMPOH, BbIMVOR, SpIMVOR, EcIMPDH and JjIMPDH should all bind tiazofurin with similar affinity, yet the values OfX 1 vary from 1-69 mM.
  • the resulting "intrinsic values” are indeed nearly identical, ranging from 0.3-0.7 mM.
  • the intrinsic values Of X 1 for ADP range from 0.2-9 mM, reflecting the structural divergence of the ADP binding sites.
  • the intrinsic affinities of A-H We determined the intrinsic values of IC50 for A-H in order to assess how competition with the mobile flap contributes to susceptibility (Figure 67). Inspection of the intrinsic values of IC 50 reveals two distinct inhibitor binding modes. The intrinsic values of IC 50 of C range between 0.18-0.36 ⁇ M for CpIMPDH, HpIMPDH, BbIMVOR and S250A/L444Y ( Figure 67), reflecting the conservation of this binding site. Likewise, the intrinsic affinities of compounds A, B, D, E and F are within a factor of 2 for all four enzymes, indicating that the binding sites of these compounds are also conserved. These observations suggest that compounds A, B, D, E and F most likely occupy the same binding site as C.
  • A-F for SpIMVOR is very different from CpIMDPH, indicating that this binding site is significantly different in SpIMVOR.
  • Met326 is a Leu in SpIMVOR.
  • the Leu substitution is also present in HpIMPDH and BbIMVOR, and therefore cannot account for the different susceptibility.
  • the next nearest substitution is Thr for Serl64; the side chain of Serl64 is 5 A away from C64, but might be closer to the A, B and D-F.
  • Alal65 and Tyr358 comprise a structural motif that defines enzymes susceptible to Q7IMPDH inhibitors.
  • a BLAST search reveals that these critical residues are present in IMPDHs from a wide variety of pathogenic bacteria in addition to C. parvum, B. burgdorferi and H.
  • Campylobacter lari food poisoning
  • Campylobacter jejuni food poisoning
  • Arcobacter butzleri food poisoning
  • Bacteroides capillosis abscesses
  • Fusobacterium nucleatum periodontitis, Lemierre's syndrome, skin ulcers
  • Burkholderia cenocepacia infection in Cystic Fibrosis
  • S. pneumoniae pneumonia
  • Clostridia botulinum botulism
  • Neisseria gonorrhoeae gonorrhea
  • Mycobacterium tuberculosis tuberculosis
  • Inosine 5 '-monophosphate dehydrogenase IMPDH
  • IMPDH Inosine 5 '-monophosphate dehydrogenase
  • Cryptosporidium parvum a major cause of diarrhea and malnutrition and a potential bioterrorism agent.
  • Q?IMPDH is most closely related to prokaryotic IMPDHs, suggesting that the parasite obtained its IMPDH gene via horizontal transfer.
  • Ncol site was created at the beginning of the Lt ⁇ MPDH coding sequence and the Ncol-Pstl fragment was cloned into pKK233-2 to create the plasmid pLDI, which expresses Lt ⁇ MPDH under control of the trc promoter.
  • Cultures were induced with 0.5 mM IPTG and grown overnight. Cells were harvested by centrifugation, resuspended in Buffer A, lysed by sonication and clarified by centrifugation followed by filtration through a 45 ⁇ m cellulose acetate filter.
  • Protein was applied to a Poros HS strong cation exchange resin (PerSeptive Biosystems) pre-equilibrated with 20 mM NaP 1 , pH 7.5, 1 mM DTT (Buffer B). Lt ⁇ MPDH was eluted with a gradient of 0-0.9 M NaCl. Fractions containing IMPDH activity were pooled and applied to IMP affinity resin. The column was washed with Buffer B and enzyme was eluted with Buffer B containing 0.5 M KCl, 1 mM IMP. The specific activity of the final preparation was 2.6 ⁇ moles/min-mg.
  • H. pylori and S. pyogenes guaB genes were cloned into pET28a with 6x His-tags. Bacteria were grown at 30 0 C in LB medium containing 25 ⁇ g/mL kanamycin until the OD OOO reached approximately 0.6. Expression was initiated by the addition of 0.5 mM IPTG and the temperature was changed to 25°C. Bacteria were harvested after 16 hours. The cell pellet was rinsed (3x) with 50 mM phosphate buffer, 500 mM NaCl, 5 mM imidazole, pH 8.0, 1 mM IMP and 5 mM ⁇ -mercaptoethanol, and lysed by sonication.
  • the lysate was clarified by centrifugation and loaded on a Ni-NTA column (Qiagen).
  • the purified protein were eluted in 50 mM phosphate buffer, 500 mM NaCl, 250 mM imidazole, pH 8.0, 1 mM IMP and 5 mM ⁇ - mercaptoethanol, concentrated and dialyzed against 50 mM Tris-HCl, pH 8.0, and 10 % glycerol.
  • the protein concentration was determined by using Bradford dye procedure (BioRad).
  • U 1 W(I + [I]/ IC 50 ) (c) where D 1 is initial velocity in the presence of inhibitor (I) and ⁇ 0 is the initial velocity in the absence of inhibitor.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Health & Medical Sciences (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Immunology (AREA)
  • Engineering & Computer Science (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Rheumatology (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

La présente invention concerne des composés, et des sels pharmaceutiquement acceptables et des promédicaments de ceux-ci, qui sont utiles en tant qu'inhibiteurs d'IMPDH. Dans certains modes de réalisation, un composé de l'invention inhibe sélectivement un IMPDH parasitaire par rapport à un IMPDH hôte. De plus, l'invention concerne des compositions pharmaceutiques comprenant un ou plusieurs composés de l'invention. L'invention concerne en outre des procédés de traitement de différentes infections parasitaires et bactériennes chez des mammifères. De plus, les composés peuvent être utilisés seuls ou en combinaison avec d'autres agents thérapeutiques ou prophylactiques, tels que des antiviraux, des agents anti-inflammatoires, des antimicrobiens et des immunosuppresseurs.
PCT/US2010/028178 2009-03-20 2010-03-22 Composés et procédés pour traiter les infections microbiennes gastro-intestinales mammaliennes WO2010108187A2 (fr)

Priority Applications (3)

Application Number Priority Date Filing Date Title
US13/257,418 US8969342B2 (en) 2009-03-20 2010-03-22 Compounds and methods for treating mammalian gastrointestinal microbial infections
EP10754239A EP2408753A4 (fr) 2009-03-20 2010-03-22 Composés et procédés pour traiter les infections microbiennes gastro-intestinales mammaliennes
US14/571,673 US10125116B2 (en) 2009-03-20 2014-12-16 Compounds and methods for treating mammalian gastrointestinal microbial infections

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US16201309P 2009-03-20 2009-03-20
US61/162,013 2009-03-20

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US13/257,418 A-371-Of-International US8969342B2 (en) 2009-03-20 2010-03-22 Compounds and methods for treating mammalian gastrointestinal microbial infections
US14/571,673 Division US10125116B2 (en) 2009-03-20 2014-12-16 Compounds and methods for treating mammalian gastrointestinal microbial infections

Publications (2)

Publication Number Publication Date
WO2010108187A2 true WO2010108187A2 (fr) 2010-09-23
WO2010108187A3 WO2010108187A3 (fr) 2011-03-31

Family

ID=42740276

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2010/028178 WO2010108187A2 (fr) 2009-03-20 2010-03-22 Composés et procédés pour traiter les infections microbiennes gastro-intestinales mammaliennes

Country Status (3)

Country Link
US (2) US8969342B2 (fr)
EP (1) EP2408753A4 (fr)
WO (1) WO2010108187A2 (fr)

Cited By (43)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011120604A1 (fr) * 2010-03-30 2011-10-06 Pharmeste S.R.L. Antagonistes du récepteur vanilloïde trpv1 ayant une partie bicyclique
ITMI20102191A1 (it) * 2010-11-25 2012-05-26 Heidelberg Inst For Theoretical Studies Hits Uso di inibitori della pteridina reduttasi per la prevenzione e/o il trattamento di infezioni parassitarie
WO2013078413A1 (fr) * 2011-11-22 2013-05-30 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Modulateurs du stockage lipidique
US8592471B2 (en) 2009-05-07 2013-11-26 Gruenenthal Gmbh Substituted phenylureas and phenylamides as vanilloid receptor ligands
WO2014006066A1 (fr) * 2012-07-06 2014-01-09 F. Hoffmann-La Roche Ag Composés triazoles en tant qu'antiviraux
WO2014028931A2 (fr) * 2012-08-17 2014-02-20 Brandeis University Composés et procédés de traitement d'infections microbiennes gastro-intestinales de mammifère
US8865912B2 (en) 2010-10-06 2014-10-21 Glaxosmithkline Llc Benzimidazole derivatives as PI3 kinase inhibitors
US8871754B2 (en) 2012-11-19 2014-10-28 Irm Llc Compounds and compositions for the treatment of parasitic diseases
CN104311598A (zh) * 2014-10-24 2015-01-28 中国农业大学 含有1,2,3-三唑环的磷酸酯化合物及其制备方法和应用
US8946204B2 (en) 2009-05-07 2015-02-03 Gruenenthal Gmbh Substituted phenylureas and phenylamides as vanilloid receptor ligands
CN104788369A (zh) * 2015-03-17 2015-07-22 三峡大学 双吡啶基苯氧基脂肪酰胺衍生物及其医药用途
CN104817491A (zh) * 2015-03-17 2015-08-05 三峡大学 N-吡啶基-2-苯氧基脂肪酰胺及其医药用途
WO2015158908A1 (fr) * 2014-04-17 2015-10-22 Institut Pasteur Korea Composés pour traiter des infections virales
US9290485B2 (en) 2010-08-04 2016-03-22 Novartis Ag N-((6-amino-pyridin-3-yl)methyl)-heteroaryl-carboxamides
CN105440020A (zh) * 2013-12-10 2016-03-30 常州大学 具有抗肿瘤活性的1,2,3-三唑类化合物及其制备方法
US9556169B2 (en) 2012-11-19 2017-01-31 Novartis Ag Compounds and compositions for the treatment of parasitic diseases
WO2017059120A1 (fr) * 2015-09-30 2017-04-06 Gilead Sciences, Inc. Composés et combinaisons pour le traitement du vih
JP2017524019A (ja) * 2014-08-15 2017-08-24 ヤンセン ファーマシューティカルズ,インコーポレーテッド Nr2b受容体阻害剤としてのトリアゾール
CN109053701A (zh) * 2018-08-03 2018-12-21 内蒙古民族大学附属医院 牛蒡子苷元类化合物、制备方法和用途
WO2019043217A1 (fr) 2017-09-04 2019-03-07 F. Hoffmann-La Roche Ag Dihydrobenzimidazolones
WO2019132638A1 (fr) * 2017-12-28 2019-07-04 Moroccan Foundation For Advanced Science, Innovation And Research (Mascir) Molécules dérivées de thiazolylbenzimidazole contenant des triazoles et leurs utilisations pour le traitement du cancer.
US10450269B1 (en) 2013-11-18 2019-10-22 Global Blood Therapeutics, Inc. Compounds and uses thereof for the modulation of hemoglobin
US10617676B2 (en) 2016-10-06 2020-04-14 Janssen Pharmaceutica Nv Substituted 1H-imidazo[4,5-b]pyridin-2(3H)-ones and their use as GluN2B receptor modulators
KR20200071572A (ko) * 2018-12-11 2020-06-19 재단법인 경기도경제과학진흥원 중동호흡기증후군 코로나바이러스에 대한 항바이러스 조성물
US10766880B2 (en) 2016-02-10 2020-09-08 Janssen Pharmaceutica Nv Substituted 1,2,3-triazoles as NR2B-selective NMDA modulators
WO2020262996A1 (fr) * 2019-06-25 2020-12-30 (주)앰틱스바이오 Composé dérivé introduisant un groupe biphényle dans un nouvel acide aminoalcanoïque et composition pharmaceutique antifongique le comprenant
WO2021091238A1 (fr) * 2019-11-05 2021-05-14 아주대학교산학협력단 Inhibiteur à petite molécule du facteur de nécrose tumorale-alpha (tnf-alpha)
US11008302B2 (en) 2018-04-04 2021-05-18 Janssen Pharmaceutica Nv Substituted pyridine and pyrimidines and their use as GluN2B receptor modulators
US11053195B2 (en) 2013-03-15 2021-07-06 Global Blood Therapeutics, Inc. Compounds and uses thereof for the modulation of hemoglobin
US11161846B2 (en) 2019-06-14 2021-11-02 Janssen Pharmaceutica Nv Substituted pyrazolo[4,3-b]pyridines and their use as GluN2B receptor modulators
US11214563B2 (en) 2019-06-14 2022-01-04 Janssen Pharmaceutica Nv Substituted pyrazolo-pyrazines and their use as GluN2B receptor modulators
US11220492B2 (en) 2017-05-17 2022-01-11 Arcus Biosciences, Inc. Quinazoline-pyrazole derivatives for the treatment of cancer-related disorders
WO2022167819A1 (fr) * 2021-02-08 2022-08-11 Cerevance, Inc. Nouveaux composés
CN114957215A (zh) * 2022-01-13 2022-08-30 渤海大学 亚甲基桥连喹啉和1,2,3-三唑双杂环化合物及其制备方法与应用
US11447503B2 (en) 2019-06-14 2022-09-20 Janssen Pharmaceutica Nv Pyridine carbamates and their use as GLUN2B receptor modulators
US11459336B2 (en) 2019-06-14 2022-10-04 Janssen Pharmaceutica Nv Pyrazine carbamates and their use as GluN2B receptor modulators
WO2022211520A1 (fr) * 2021-04-02 2022-10-06 파렌키마바이오텍 주식회사 Nouveau composé et son utilisation pour le traitement d'une maladie auto-immune
US11530210B2 (en) 2019-06-14 2022-12-20 Janssen Pharmaceutica Nv Substituted heteroaromatic pyrazolo-pyridines and their use as GLUN2B receptor modulators
US11618750B2 (en) 2019-06-14 2023-04-04 Janssen Pharmaceutica Nv Substituted pyrazolo-pyridine amides and their use as GluN2B receptor modulators
WO2023128033A1 (fr) * 2021-12-31 2023-07-06 (주)앰틱스바이오 Nouvelle composition antifongique pour inhiber la production de biofilm de champignons infectieux
US11820747B2 (en) 2021-11-02 2023-11-21 Flare Therapeutics Inc. PPARG inverse agonists and uses thereof
WO2024077036A1 (fr) * 2022-10-07 2024-04-11 Scorpion Therapeutics, Inc. Méthodes de traitement du cancer
US12030879B2 (en) 2018-03-02 2024-07-09 Inflazome Limited Sulfonyl acetamides as NLRP3 inhibitors

Families Citing this family (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8969342B2 (en) 2009-03-20 2015-03-03 Brandeis University Compounds and methods for treating mammalian gastrointestinal microbial infections
WO2013012848A1 (fr) 2011-07-18 2013-01-24 Merck Patent Gmbh Benzamides
KR101435496B1 (ko) * 2012-10-22 2014-08-28 한국과학기술연구원 미토콘드리아 기능 조절제로서의 벤즈이미다졸 유도체
US9246108B2 (en) 2012-12-28 2016-01-26 Dow Global Technologies Llc Quinoline-benzoxazole derived compounds for electronic films and devices
US9981923B2 (en) 2014-06-20 2018-05-29 Council Of Scientific & Industrial Research 1,2,3 Triazole antifungal agents and preparation thereof
WO2016007837A1 (fr) * 2014-07-11 2016-01-14 Spero Therapeutics, Inc. Inhibiteurs de la tolérance aux antibiotiques à base d'hétéroaryle bicyclique à liaison carbonyle
WO2020112605A1 (fr) * 2018-11-30 2020-06-04 Brandeis University Composés et méthodes de traitement d'infections microbiennes chroniques
US20230338350A1 (en) * 2020-07-03 2023-10-26 Nihon Nohyaku Co., Ltd. Anticoccidial agent and method for using the same

Family Cites Families (25)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE2200325C2 (de) 1971-12-31 1986-10-30 Showa Denko K.K., Tokio/Tokyo Verwendung von 3-(1-Phenylisopropyl)-harnstoff-Derivaten als Herbizide
AU464662B2 (en) 1971-12-31 1975-09-04 Showa Denko Kabushiki Kaisha Novel 3-(2-phenylisopropyl) urea derivatives
US3931203A (en) 1973-03-19 1976-01-06 Rohm And Haas Company 3-Pyridylmethyl aryl urea rodenticides
AU506399B2 (en) 1975-12-30 1980-01-03 Showa Denko Kabushiki Kaisha 3-(2-aryl-2-propyl) ureas
JPH0929260A (ja) * 1995-07-24 1997-02-04 Daikin Ind Ltd 水処理装置
US5807876A (en) 1996-04-23 1998-09-15 Vertex Pharmaceuticals Incorporated Inhibitors of IMPDH enzyme
SK286662B6 (sk) 1996-04-23 2009-03-05 Vertex Pharmaceuticals Incorporated Deriváty močoviny, farmaceutické prostriedky, ktoré ich obsahujú, a ich použitie ako inhibítorov aktivity IMPDH enzýmu
AU8534198A (en) 1997-08-05 1999-03-01 Novo Nordisk A/S Derivatives of 2,5- and 3,5-disubstituted anilines, their preparation and use
AU1455500A (en) * 1998-10-29 2000-05-22 Trega Biosciences, Inc. Oxadiazole, thiadiazole and triazole derivatives and combinatorial libraries thereof
US6867299B2 (en) 2000-02-24 2005-03-15 Hoffmann-La Roche Inc. Oxamide IMPDH inhibitors
PE20030701A1 (es) * 2001-12-20 2003-08-21 Schering Corp Compuestos para el tratamiento de trastornos inflamatorios
US6914058B2 (en) * 2002-01-18 2005-07-05 Dr. Reddy's Laboratories, Limited Antibacterial compounds: process for their preparation and pharmaceutical compositions containing them
US7317030B2 (en) 2004-02-26 2008-01-08 4Sc Ag Compounds as inhibitors of cell proliferation and viral infections
WO2007008529A2 (fr) 2005-07-08 2007-01-18 Kalypsys, Inc Agents modificateurs de l'absorption du cholesterol cellulaire
AR057455A1 (es) * 2005-07-22 2007-12-05 Merck & Co Inc Inhibidores de la transcriptasa reversa de vih y composicion farmaceutica
EP1986633B1 (fr) * 2006-02-10 2014-07-30 Summit Corporation Plc Traitement de la dystrophie musculaire de duchenne
JP4986485B2 (ja) * 2006-03-28 2012-07-25 株式会社Adeka エポキシ樹脂硬化性組成物
WO2007143557A2 (fr) 2006-06-02 2007-12-13 Brandeis University Composés et procédés de traitement des infections parasitaires gastro-intestinales des mammifères
US8455513B2 (en) * 2007-01-10 2013-06-04 Aerie Pharmaceuticals, Inc. 6-aminoisoquinoline compounds
WO2009018344A1 (fr) 2007-07-30 2009-02-05 Regents Of The University Of Minnesota Agents anticancéreux
AR068332A1 (es) * 2007-08-03 2009-11-11 Summit Corp Plc Combinaciones de drogas para el tratamiento de la distrofia muscular de duchenne
US20110177999A1 (en) * 2007-08-09 2011-07-21 Vertex Pharmaceuticals Incorporated Therapeutic Combinations Useful in Treating CFTR Related Diseases
BRPI0912539A2 (pt) 2008-05-05 2015-10-13 Amgen Inc composto, composição farmacêutica , método para tratar uma doença, e, uso do composto.
US8969342B2 (en) 2009-03-20 2015-03-03 Brandeis University Compounds and methods for treating mammalian gastrointestinal microbial infections
US9447134B2 (en) 2012-08-17 2016-09-20 Brandeis University Compounds and methods for treating mammalian gastrointestinal microbial infections

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of EP2408753A4 *

Cited By (76)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8946204B2 (en) 2009-05-07 2015-02-03 Gruenenthal Gmbh Substituted phenylureas and phenylamides as vanilloid receptor ligands
US8592471B2 (en) 2009-05-07 2013-11-26 Gruenenthal Gmbh Substituted phenylureas and phenylamides as vanilloid receptor ligands
US9624209B2 (en) 2009-05-07 2017-04-18 Gruenenthal Gmbh Substituted phenylureas and phenylamides as vanilloid receptor ligands
US9120756B2 (en) 2009-05-07 2015-09-01 Gruenenthal Gmbh Substituted phenylureas and phenylamides as vanilloid receptor ligands
US9216975B2 (en) 2010-03-30 2015-12-22 Serentrix, LLC. TRPV1 vanilloid receptor antagonists with a bicyclic portion
WO2011120604A1 (fr) * 2010-03-30 2011-10-06 Pharmeste S.R.L. Antagonistes du récepteur vanilloïde trpv1 ayant une partie bicyclique
US9290485B2 (en) 2010-08-04 2016-03-22 Novartis Ag N-((6-amino-pyridin-3-yl)methyl)-heteroaryl-carboxamides
US9156797B2 (en) 2010-10-06 2015-10-13 Glaxosmithkline Llc Benzimidazole derivatives as PI3 kinase inhibitors
US10314845B2 (en) 2010-10-06 2019-06-11 Glaxosmithkline Llc Benzimidazole derivatives as PI3 kinase inhibitors
US9872860B2 (en) 2010-10-06 2018-01-23 Glaxosmithkline Llc Benzimidazole derivatives as PI3 kinase inhibitors
US8865912B2 (en) 2010-10-06 2014-10-21 Glaxosmithkline Llc Benzimidazole derivatives as PI3 kinase inhibitors
US9062003B2 (en) 2010-10-06 2015-06-23 Glaxosmithkline Llc Benzimidazole derivatives as PI3 kinase inhibitors
US10660898B2 (en) 2010-10-06 2020-05-26 Glaxosmithkline Llc Benzimidazole derivatives as PI3 kinase inhibitors
ITMI20102191A1 (it) * 2010-11-25 2012-05-26 Heidelberg Inst For Theoretical Studies Hits Uso di inibitori della pteridina reduttasi per la prevenzione e/o il trattamento di infezioni parassitarie
WO2013078413A1 (fr) * 2011-11-22 2013-05-30 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Modulateurs du stockage lipidique
WO2014006066A1 (fr) * 2012-07-06 2014-01-09 F. Hoffmann-La Roche Ag Composés triazoles en tant qu'antiviraux
WO2014028931A2 (fr) * 2012-08-17 2014-02-20 Brandeis University Composés et procédés de traitement d'infections microbiennes gastro-intestinales de mammifère
WO2014028931A3 (fr) * 2012-08-17 2014-10-09 Brandeis University Composés et procédés de traitement d'infections microbiennes gastro-intestinales de mammifère
US9447134B2 (en) 2012-08-17 2016-09-20 Brandeis University Compounds and methods for treating mammalian gastrointestinal microbial infections
US8871754B2 (en) 2012-11-19 2014-10-28 Irm Llc Compounds and compositions for the treatment of parasitic diseases
US9926314B2 (en) 2012-11-19 2018-03-27 Novartis Ag Compounds and compositions for the treatment of parasitic diseases
US9556169B2 (en) 2012-11-19 2017-01-31 Novartis Ag Compounds and compositions for the treatment of parasitic diseases
US11053195B2 (en) 2013-03-15 2021-07-06 Global Blood Therapeutics, Inc. Compounds and uses thereof for the modulation of hemoglobin
US10450269B1 (en) 2013-11-18 2019-10-22 Global Blood Therapeutics, Inc. Compounds and uses thereof for the modulation of hemoglobin
CN105440020A (zh) * 2013-12-10 2016-03-30 常州大学 具有抗肿瘤活性的1,2,3-三唑类化合物及其制备方法
CN105440020B (zh) * 2013-12-10 2018-12-18 常州大学 具有抗肿瘤活性的1,2,3-三唑类化合物及其制备方法
US9809602B2 (en) 2014-04-17 2017-11-07 Institut Pasteur Korea Compounds for treating viral infections
KR102346605B1 (ko) * 2014-04-17 2021-12-31 재단법인 한국파스퇴르연구소 바이러스성 감염 치료용 화합물
KR20160141839A (ko) * 2014-04-17 2016-12-09 재단법인 한국파스퇴르연구소 바이러스성 감염 치료용 화합물
JP2017515891A (ja) * 2014-04-17 2017-06-15 インスティチュート パストゥール コリア ウイルス性感染治療用化合物
WO2015158908A1 (fr) * 2014-04-17 2015-10-22 Institut Pasteur Korea Composés pour traiter des infections virales
JP2017524019A (ja) * 2014-08-15 2017-08-24 ヤンセン ファーマシューティカルズ,インコーポレーテッド Nr2b受容体阻害剤としてのトリアゾール
CN104311598A (zh) * 2014-10-24 2015-01-28 中国农业大学 含有1,2,3-三唑环的磷酸酯化合物及其制备方法和应用
CN104817491A (zh) * 2015-03-17 2015-08-05 三峡大学 N-吡啶基-2-苯氧基脂肪酰胺及其医药用途
CN104817491B (zh) * 2015-03-17 2017-06-06 三峡大学 N‑吡啶基‑2‑苯氧基脂肪酰胺及其医药用途
CN104788369B (zh) * 2015-03-17 2017-06-06 三峡大学 双吡啶基苯氧基脂肪酰胺衍生物及其医药用途
CN104788369A (zh) * 2015-03-17 2015-07-22 三峡大学 双吡啶基苯氧基脂肪酰胺衍生物及其医药用途
WO2017059120A1 (fr) * 2015-09-30 2017-04-06 Gilead Sciences, Inc. Composés et combinaisons pour le traitement du vih
JP2018529714A (ja) * 2015-09-30 2018-10-11 ギリアード サイエンシーズ, インコーポレイテッド Hivの処置のための化合物および組合せ物
US10059697B2 (en) 2015-09-30 2018-08-28 Gilead Sciences, Inc. Compounds and combinations for the treatment of HIV
US10766880B2 (en) 2016-02-10 2020-09-08 Janssen Pharmaceutica Nv Substituted 1,2,3-triazoles as NR2B-selective NMDA modulators
US11207298B2 (en) 2016-10-06 2021-12-28 Janssen Pharmaceutica Nv Substituted 1H-imidazo[4,5-b]pyridin-2(3H)-ones and their use as GLUN2B receptor modulators
US10617676B2 (en) 2016-10-06 2020-04-14 Janssen Pharmaceutica Nv Substituted 1H-imidazo[4,5-b]pyridin-2(3H)-ones and their use as GluN2B receptor modulators
US11759455B2 (en) 2016-10-06 2023-09-19 Janssen Pharmaceutica Nv Substituted 1H-imidazo[4,5-b]pyridin-2(3H)-ones and their use as GLUN2B receptor modulators
US11220492B2 (en) 2017-05-17 2022-01-11 Arcus Biosciences, Inc. Quinazoline-pyrazole derivatives for the treatment of cancer-related disorders
WO2019043217A1 (fr) 2017-09-04 2019-03-07 F. Hoffmann-La Roche Ag Dihydrobenzimidazolones
US11254672B2 (en) 2017-09-04 2022-02-22 C4 Therapeutics, Inc. Dihydrobenzimidazolones for medical treatment
US11787802B2 (en) 2017-09-04 2023-10-17 C4 Therapeutics, Inc. Dihydrobenzimidazolones for medical treatment
WO2019132638A1 (fr) * 2017-12-28 2019-07-04 Moroccan Foundation For Advanced Science, Innovation And Research (Mascir) Molécules dérivées de thiazolylbenzimidazole contenant des triazoles et leurs utilisations pour le traitement du cancer.
US12030879B2 (en) 2018-03-02 2024-07-09 Inflazome Limited Sulfonyl acetamides as NLRP3 inhibitors
US11008302B2 (en) 2018-04-04 2021-05-18 Janssen Pharmaceutica Nv Substituted pyridine and pyrimidines and their use as GluN2B receptor modulators
CN109053701A (zh) * 2018-08-03 2018-12-21 内蒙古民族大学附属医院 牛蒡子苷元类化合物、制备方法和用途
CN109053701B (zh) * 2018-08-03 2020-06-23 内蒙古民族大学附属医院 牛蒡子苷元类化合物、制备方法和用途
KR20200071572A (ko) * 2018-12-11 2020-06-19 재단법인 경기도경제과학진흥원 중동호흡기증후군 코로나바이러스에 대한 항바이러스 조성물
KR102135106B1 (ko) 2018-12-11 2020-07-17 재단법인 경기도경제과학진흥원 중동호흡기증후군 코로나바이러스에 대한 항바이러스 조성물
US11447503B2 (en) 2019-06-14 2022-09-20 Janssen Pharmaceutica Nv Pyridine carbamates and their use as GLUN2B receptor modulators
US11161846B2 (en) 2019-06-14 2021-11-02 Janssen Pharmaceutica Nv Substituted pyrazolo[4,3-b]pyridines and their use as GluN2B receptor modulators
US11993587B2 (en) 2019-06-14 2024-05-28 Janssen Pharmaceutica Nv Substituted pyrazolo-pyrazines and their use as GluN2B receptor modulators
US11214563B2 (en) 2019-06-14 2022-01-04 Janssen Pharmaceutica Nv Substituted pyrazolo-pyrazines and their use as GluN2B receptor modulators
US11618750B2 (en) 2019-06-14 2023-04-04 Janssen Pharmaceutica Nv Substituted pyrazolo-pyridine amides and their use as GluN2B receptor modulators
US11530210B2 (en) 2019-06-14 2022-12-20 Janssen Pharmaceutica Nv Substituted heteroaromatic pyrazolo-pyridines and their use as GLUN2B receptor modulators
US11459336B2 (en) 2019-06-14 2022-10-04 Janssen Pharmaceutica Nv Pyrazine carbamates and their use as GluN2B receptor modulators
CN114051496A (zh) * 2019-06-25 2022-02-15 阿米蒂比奥有限公司 将联苯基引入新型氨基烷酸的衍生物化合物及包含其的抗真菌药学组合物
KR20210030343A (ko) * 2019-06-25 2021-03-17 (주)앰틱스바이오 신규한 아미노알칸산에 바이페닐기를 도입한 유도체 화합물 및 이를 포함하는 항진균성 약학적 조성물
US11479526B2 (en) 2019-06-25 2022-10-25 Amtixbio Co., Ltd. Aminoalkanoic acid derivative containing biphenyl group and antifungal pharmaceutical composition comprising the same
JP2022535613A (ja) * 2019-06-25 2022-08-09 エムティックスバイオ カンパニー リミテッド 新規なアミノアルカン酸にビフェニル基を導入した誘導体化合物、およびこれを含む抗真菌薬学的組成物
JP7290361B2 (ja) 2019-06-25 2023-06-13 エムティックスバイオ カンパニー リミテッド 新規なアミノアルカン酸にビフェニル基を導入した誘導体化合物、およびこれを含む抗真菌薬学的組成物
KR102286897B1 (ko) 2019-06-25 2021-08-09 (주)앰틱스바이오 신규한 아미노알칸산에 바이페닐기를 도입한 유도체 화합물 및 이를 포함하는 항진균성 약학적 조성물
WO2020262996A1 (fr) * 2019-06-25 2020-12-30 (주)앰틱스바이오 Composé dérivé introduisant un groupe biphényle dans un nouvel acide aminoalcanoïque et composition pharmaceutique antifongique le comprenant
WO2021091238A1 (fr) * 2019-11-05 2021-05-14 아주대학교산학협력단 Inhibiteur à petite molécule du facteur de nécrose tumorale-alpha (tnf-alpha)
WO2022167819A1 (fr) * 2021-02-08 2022-08-11 Cerevance, Inc. Nouveaux composés
WO2022211520A1 (fr) * 2021-04-02 2022-10-06 파렌키마바이오텍 주식회사 Nouveau composé et son utilisation pour le traitement d'une maladie auto-immune
US11820747B2 (en) 2021-11-02 2023-11-21 Flare Therapeutics Inc. PPARG inverse agonists and uses thereof
WO2023128033A1 (fr) * 2021-12-31 2023-07-06 (주)앰틱스바이오 Nouvelle composition antifongique pour inhiber la production de biofilm de champignons infectieux
CN114957215A (zh) * 2022-01-13 2022-08-30 渤海大学 亚甲基桥连喹啉和1,2,3-三唑双杂环化合物及其制备方法与应用
WO2024077036A1 (fr) * 2022-10-07 2024-04-11 Scorpion Therapeutics, Inc. Méthodes de traitement du cancer

Also Published As

Publication number Publication date
EP2408753A2 (fr) 2012-01-25
US20150099781A1 (en) 2015-04-09
WO2010108187A3 (fr) 2011-03-31
US10125116B2 (en) 2018-11-13
US20120101096A1 (en) 2012-04-26
EP2408753A4 (fr) 2012-11-07
US8969342B2 (en) 2015-03-03

Similar Documents

Publication Publication Date Title
US10125116B2 (en) Compounds and methods for treating mammalian gastrointestinal microbial infections
US9447134B2 (en) Compounds and methods for treating mammalian gastrointestinal microbial infections
USRE47428E1 (en) Modulators of methyl modifying enzymes, compositions and uses thereof
EP2953941B1 (fr) Modulateurs d'enzymes de modification par méthylation, compositions et utilisations associées
JP2020524158A (ja) Ssao阻害剤
EP3634958B1 (fr) Agonistes du récepteur 2 du peptide formylé et agonistes du récepteur 1 du peptide formylé à base de cyclopropyle d'urée
EP2734516B1 (fr) Éthers d'arylpyrazole en tant qu'inhibiteurs de leucotriène a4 hydrolase
KR20210143775A (ko) 베타 아드레날린성 작용제 및 이의 사용 방법
US20120264760A1 (en) Compounds and Methods for Treating Mammalian Gastrointestinal Parasitic Infections
EA004141B1 (ru) Ингибиторы фермента impdh
CA2888360A1 (fr) Composes et compositions therapeutiques
US10745405B2 (en) Glucuronide prodrugs of Janus kinase inhibitors
EP3417850A1 (fr) Dérivés de benzotriazole trisubstitués à titre d'inhibiteurs de dihydroorotate oxygénase
IL258225A (en) Ppar agonists, compounds, pharmaceuticals and methods of using them
JP2019509337A (ja) プラスメプシンv阻害薬としてのイミノチアジアジン二量体誘導体
KR20100016584A (ko) 암 치료제로서의 나프탈렌 카르복실산 아미드의 에테르
CZ299387B6 (cs) Antivirové slouceniny
EP4155304A1 (fr) Composé utilisé comme inhibiteur de kinase ret et son application
RU2468009C2 (ru) Ингибитор связывания s1p1
TW201902880A (zh) 使用三取代苯并三唑衍生物作為二氫乳清酸氧合酶抑制劑之方法
US11608328B2 (en) Chemical probes to identify anti-mycobacterial MmpL3 inhibitors
WO2020143800A1 (fr) Agoniste de l'ampk spécifique au foie, son procédé de préparation et ses applications
JP2024500179A (ja) ピロール誘導体、その調製方法及びその用途
KR100643057B1 (ko) Impdh효소억제제로서의우레아유도체

Legal Events

Date Code Title Description
NENP Non-entry into the national phase in:

Ref country code: DE

121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 10754239

Country of ref document: EP

Kind code of ref document: A2

WWE Wipo information: entry into national phase

Ref document number: 2010754239

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 13257418

Country of ref document: US